US20070048726A1 - Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State - Google Patents

Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State Download PDF

Info

Publication number
US20070048726A1
US20070048726A1 US11/553,245 US55324506A US2007048726A1 US 20070048726 A1 US20070048726 A1 US 20070048726A1 US 55324506 A US55324506 A US 55324506A US 2007048726 A1 US2007048726 A1 US 2007048726A1
Authority
US
United States
Prior art keywords
gel
solution
cell
agent
preservation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/553,245
Inventor
John Baust
Robert Van Buskirk
Kristi Snyder
Aby Mathew
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biolife Solutions Inc
Original Assignee
Biolife Solutions Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/757,694 external-priority patent/US6632666B2/en
Application filed by Biolife Solutions Inc filed Critical Biolife Solutions Inc
Priority to US11/553,245 priority Critical patent/US20070048726A1/en
Assigned to BIOLIFE SOLUTIONS, INC. reassignment BIOLIFE SOLUTIONS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAUST, JOHN G, BAUST, JOHN M, MATHEW, ABY J, SNYDER, KRISTI K, VAN BUSKIRK, ROBERT
Publication of US20070048726A1 publication Critical patent/US20070048726A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0226Physiologically active agents, i.e. substances affecting physiological processes of cells and tissue to be preserved, e.g. anti-oxidants or nutrients
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/0231Chemically defined matrices, e.g. alginate gels, for immobilising, holding or storing cells, tissue or organs for preservation purposes; Chemically altering or fixing cells, tissue or organs, e.g. by cross-linking, for preservation purposes

Definitions

  • the invention relates to the gel state preservation of cells, tissues and organs.
  • the invention relates to the hypothermic preservation or storage of cells, tissues or organs in the semi-solid, gel-like state.
  • preservation of biologics (cells, tissues, and organs) has traditionally been achieved through “suspension” in a liquid preservation medium.
  • liquid preservation medium include, but are not limited to a simple saline solution, cell culture media, and preservation solutions such as University of Wisconsin (UW) solution (Viaspan® solution), Euro-CollinsTM solution, and Hypothermosol® solution (Bio Life Solutions, Inc., Owego, N.Y., USA).
  • UW University of Wisconsin
  • Vertex® solution Very-CollinsTM solution
  • Hypothermosol® solution Bio Life Solutions, Inc., Owego, N.Y., USA.
  • Inherent in this liquid preservation approach is that the liquid environment confers no physical support network for the biologic during preservation and transport. Due to this lack of physical support upon preservation, biologics are exposed to numerous physical stresses during storage and shipment.
  • hypothermic preservation The preservation of eukaryotic cells, tissues or organs is commonly carried out by chilling them sufficiently to slow or halt metabolic processes that require ongoing support by an organism or the environment to maintain viability.
  • the preservation of cells, tissues or organs by such chilling is generally referred to as hypothermic preservation or hypothermic storage.
  • hypothermic storage for cells, tissues or organs.
  • the first involves storage at temperatures above the freezing point of the solution or medium in which the sample is suspended or immersed or with which the sample is perfused.
  • temperatures may be in the range of about 10° C. to 0° C.
  • Such conditions are appropriate only for short term storage, generally on the order of hours to several days to about a week.
  • the second broad type involves storage at lower temperatures, for example, as low as ⁇ 80° C. to ⁇ 196° C. Storage under these conditions is more appropriate for longer periods of time.
  • the first sub-class involves the freezing of the samples in a medium or solution that permits the formation of ice crystals.
  • cryoprotective agents e.g., dimethyl sulfoxide (DMSO), glycerol
  • DMSO dimethyl sulfoxide
  • glycerol e.g., dimethyl sulfoxide
  • This approach is referred to herein as “cryopreservation.”
  • hypothermic storage The basic challenge of hypothermic storage is to preserve the material in a state that can be reversed without causing extensive cell damage or cell death.
  • Approaches (solutions and methods) that minimize the accumulation of sub-lethal damages such as oncotic balance, energy deprivation, cellular waste accumulation, ionic balance, as well as formation of ice crystals in or around cells are well known to aid in the survival and ultimate recovery of material stored at hypothermic temperatures.
  • sub-lethal damages such as oncotic balance, energy deprivation, cellular waste accumulation, ionic balance, as well as formation of ice crystals in or around cells.
  • ionic balance ionic balance
  • formation of ice crystals in or around cells are well known to aid in the survival and ultimate recovery of material stored at hypothermic temperatures.
  • cell death is known to occur by two different mechanisms. The first, necrotic cell death or necrosis, is not mediated by a specific cellular pathway.
  • necrosis is characterized by the loss of cell membrane integrity resulting in cell swelling, and is caused by a number of pathological agents. DNA in cells that undergo necrosis is cleaved in a random fashion. Thus, the DNA from cells that have undergone necrosis appears as a continuous smear when subjected to gel electrophoresis.
  • the second cell death mechanism, apoptosis or programmed cell death is the result of the activation of a specific biochemical pathway involving a cascade of biochemical activation steps that ultimately result in the death of the cell.
  • Apoptosis is characterized by cell shrinkage, intact plasma membranes, and non-random cleavage of DNA at an approximately 180 nucleotide interval, evidenced by a ladder of DNA cleavage products upon gel electrophoresis of genomic DNA. Apoptosis is reviewed, for example, by Kerr et al., 1994, Cancer 73: 2013 and Evan & Littlewood, 1998, Science 281: 1317.
  • U.S. Pat. No. 6,921,633 discloses methods for the hypothermic preservation of cells, tissues and organs in the vitreous state. This preservation can only be accomplished at very low temperatures. Preservation of cells, tissues, and organs in a vitreous state requires the utilization of high concentrations of toxic agents coupled with controlled reduction of sample temperature to ultra-low temperatures ( ⁇ 140° C.) to cause the formation of a glass (amorphous solid). The extent of cell survival following vitreous preservations can be significantly improved by the inclusion of anti-apoptotic agents into the preservation medium. This patent is incorporated herein by reference.
  • the present invention includes methods and compositions for the preservation of cells, tissues or organs in the semi-solid state.
  • the invention includes gel-based medium compositions for normothermic, hypothermic or cryopreservative transport and/or storage of plant tissues or cells and animal organs, tissues or cells.
  • the gel-based compositions include a cell maintenance and preservation medium and a gelling agent.
  • mammalian samples such as human and animal organs, tissues and cells, may be preserved in the gel-based media compositions.
  • the cell maintenance and preservation medium is liquid.
  • a method of preserving a eukaryotic cell, tissue or organ includes the step of contacting the cell, tissue or organ with a semi-solid, gel-like storage solution.
  • the storage solution preferably includes a composition that inhibits the activation and progression of molecular-based cell death cascades (apoptosis and necrosis) and a concentration of a semi-solid gelling agent that is sufficient for gelling of the solution.
  • the storage solution also preferably includes agents which protect cells from ice-related damage (including but not limited to dimethyl sulfoxide, glycerol, and propanediol).
  • the method also includes the step of preserving the cell, tissue or organ, where the cell, tissue or organ becomes encapsulated within the semi-solid gel-state preservation matrix.
  • the molecular based cell death control during gel-state preservation is preferably accomplished through use of a hypothermic storage solution which contains a gelling agent and agents designed to inhibit the activation and progression of apoptotic and necrotic cell death cascades.
  • a hypothermic storage solution which contains a gelling agent and agents designed to inhibit the activation and progression of apoptotic and necrotic cell death cascades.
  • the inclusion of one or more anti-apoptotic agents aids in preventing the apoptotic cell death that normally occurs subsequent to this type of preservation.
  • the gel-state preservation is performed at a temperature of +37° C. to ⁇ 0° C.
  • the gel-state preservation is performed at a temperature of ⁇ 1° C. to ⁇ 196° C., and more preferably at a temperature of ⁇ 80° C. to ⁇ 196° C.
  • the composition that inhibits apoptosis includes an agent that interacts with a polypeptide that participates in an apoptotic pathway. In one embodiment, the agent inhibits the activity of the polypeptide. In another embodiment, the agent maintains or potentiates the activity of the polypeptide.
  • the agent is selected from the group consisting of a caspase inhibitor, a calpain inhibitor, and an inhibitor of nitrous oxide synthase.
  • the composition that inhibits apoptosis includes an antioxidant.
  • the composition that inhibits apoptosis includes an agent selected from the group consisting of a free radical scavenger, a zinc chelator, and a calcium chelator.
  • FIG. 1 graphically depicts post-storage recovery of MDCK (Madin Darby Canine Kidney) cells stored as monolayers in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • MDCK Medin Darby Canine Kidney
  • FIG. 2 graphically depicts post-storage recovery of MDCK cells stored as monolayers in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 3 graphically depicts post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 4 graphically depicts post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 5 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 1 day in maintenance and preservation solutions as gel formulations.
  • FIG. 6 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 3 days in maintenance and preservation solutions as gel formulations.
  • FIG. 7 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 8° C. for 1 day in maintenance and preservation solutions as gel formulations.
  • FIG. 8 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans stored in suspension in culture plates at 8° C. for 3 days in maintenance and preservation solutions as gel formulations.
  • FIG. 9 shows post-storage function of human islets stored for 3 days at 22° C.
  • the present invention preserves cells in the gel-state (semi-solid matrix).
  • Gel-state preservation offers a number of advantages over liquidous preservation where samples can be suspended in a liquid which then solidifies, placing the specimen into a semi-solid suspended state for preservation.
  • Gel state preservation provides for a number of additional protective avenues beyond liquidous storage including, but not limited to, physical protection from sheer stress and strain, mechanical damage, specimen settling and clumping.
  • gel-state preservation offers the benefit of altering the formation and structure of ice crystals thereby reducing the negative physical effects of sub-freezing specimen storage.
  • gel-state means establishing a semi-solid matrix state in a solution and in cells, tissue or organs suspended in or perfused with that solution.
  • a “gel-state” is a semi-solid formed from a liquid without the formation of crystals.
  • a gel-state refers more particularly to a solid formed from a liquid without the formation of ice crystals. Gel-state preservation is accomplished by reducing the temperature of a solution below the gel solidification point, a variable temperature point based on specific composition ranging from 37° C. to ⁇ 196° C., such that the cells, tissue or organs are suspended in or perfused with that solution. That is, “gel-state,” as it is used herein, is when a cell, tissue or organ is encapsulated in a semisolid matrix (i.e., in the hypothermic storage solution).
  • Hypothermic storage solution refers to a solution in which cells, tissues, or organs can be stored at temperatures below physiological temperature. Preferred hypothermic storage solutions are described below.
  • composition can have one or more component elements.
  • the term “inhibit” means to reduce an activity by at least 10%, and preferably more, e.g., 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more, up to and including 100% relative to that activity that is not subject to such inhibition.
  • an agent that inhibits apoptosis inhibits apoptosis and/or necrosis by at least 10% relative to a sample subject to the same apoptotic stimulus but absent the agent.
  • the term “enhance” means to increase an activity by at least 10%, and preferably more, e.g., 20%, 50%, 75% or even 100% or more (e.g., 2 ⁇ , 5 ⁇ , 10 ⁇ , etc.) relative to that activity that is not subject to such enhancement.
  • an “anti-apoptotic agent” refers to a composition that inhibits apoptosis and/or necrosis triggered by or accompanying gel-state preservation of a cell, tissue or organ.
  • Anti-apoptotic agents fall into two broad classes, those that interact with a polypeptide that participates in an apoptotic pathway (e.g., participation in the cellular generation, propagation or execution of an apoptotic signal) and those that inhibit by other means, for example, frequently by avoiding or countering the effects of oxidative stress that tend to activate the apoptotic program.
  • the present invention uses an anti-apoptotic agent or composition that only targets one or more polypeptide targets.
  • the present invention uses an anti-apoptotic agent or composition that only targets other aspects of the apoptotic process, e.g., changes in redox status or potential.
  • the present invention uses one or more agents or compositions that act upon either of these sites.
  • the term “maintains or potentiates the activity of a polypeptide” refers to compositions or agents that prevent a decrease or cause an increase in a given activity of a given polypeptide.
  • an agent that maintains or potentiates the activity of that polypeptide prevents a decrease in the anti-apoptotic activity of that polypeptide, thereby preventing or inhibiting apoptosis (i.e., inhibiting apoptosis by at least 10% relative to a sample not treated with that agent and subject to the same apoptotic stimulus).
  • composition that interacts with a polypeptide that participates in an apoptotic pathway refers to a composition comprising one or more agents that, alone or together, physically interact with a polypeptide target in the apoptotic pathway such that apoptosis is decreased or inhibited.
  • Non-limiting examples of such polypeptides are listed in Table I A and B.
  • polypeptide that participates in an apoptotic pathway refers to a polypeptide, at least one function of which is to promote or inhibit apoptosis.
  • one class of such polypeptides includes a polypeptide that acts to promote apoptosis—the inhibition of its function or expression, e.g., a specific protease activity, leads to an inhibition of apoptosis.
  • Another class of such polypeptides includes a polypeptide that acts to inhibit apoptosis—the activation or maintenance of its function or expression, e.g., Bcl-2 activity, also leads to an inhibition of apoptosis.
  • Hypothermic storage solutions useful for semi-solid or gel-state preservation can be readily adapted for use in the methods and compositions of the present invention. These include, but are not limited to, various tissue culture medias, saline solutions, HypoThermosol® solution, Viaspan® solution, CryoStorTM solution, Celsior® solution, HTK® solution, Euro-collinsTM solution, Normosol® solution, CardisolTM solution, Unisol® solution, and Plasmalite® solution.
  • gel-based maintenance and preservation media have been developed to sustain biologics in supporting environments of various viscosity.
  • the gel-based media are rigid or highly viscous at temperatures equal to or lower than 28° C. but liquid at 37° C.
  • Liquid maintenance and preservation media i.e., Hypothermosol® solution, Viaspan® solution, also called University of Wisconsin solution, EuroCollinsTM solution, CardisolTM solution, Unisol® solutions, tissue culture media, etc.
  • Hypothermosol® solution i.e., Hypothermosol® solution, Viaspan® solution, also called University of Wisconsin solution, EuroCollinsTM solution, CardisolTM solution, Unisol® solutions, tissue culture media, etc.
  • Suitable combinations of such liquid maintenance and preservation solutions may be used if desired.
  • Hypothermosol® solution is composed of (a) one or more electrolytes selected from the group consisting of potassium ions at a concentration ranging from about 10-145 mM, sodium ions ranging from about 10-120 mM, magnesium ions ranging from about 0.1-10 mM, and calcium ions ranging from about 0.01-1.0 mM, (b) a macromolecular oncotic agent having a size sufficiently large to limit escape from the circulation system and effective to maintain oncotic pressure equivalent to that of blood plasma and selected from the group consisting of human serum albumin, polysaccharide and colloidal starch, (c) a biological pH buffer effective under physiological and hypothermic conditions, (d) a nutritive effective amount of at least one simple sugar, (e) an impermeant and hydroxyl radical scavenging effective amount of mannitol, (f) an impermeant anion impermeable to cell membranes and effective to counteract cell swelling during cold exposure, the imper
  • Adjusting the formulation of 1) the carrier medium, in particular, a cell maintenance and preservation solution, 2) gel-type and 3) gel concentration produces an appropriate long-term (up to two weeks) shipping environment, which is commercially known as GELSTOR® solution (Biolife Solutions, Inc., Owego, N.Y., USA).
  • GELSTOR® solution Biolife Solutions, Inc., Owego, N.Y., USA.
  • a biologic of choice is exposed to liquefied gel at 37° C., and thereafter cooled to a desired preservation temperature for maintenance and shipment. Upon arrival or point-of-use, the gel is warmed to 37° C. whereupon it liquefies, and may be decanted. The biologic may then be rinsed with an appropriate medium and is ready for use.
  • gel-based preservation and maintenance technology provides a semi-solid preservation matrix that facilitates protection of various cells from mechanical stresses associated with biologic preservation and transport.
  • gel-based preservation media provide physiological and biochemical protection during the storage and transport of cells, organs and tissues.
  • a variety of animal and plant cells may be stored and transported in gel-based media, including human cells and animals cells from numerous tissue sources, including, but not limited to, tumor cells, cells from the liver, kidney or central nervous system, epidermal keratinocytes, endothelial cells, stem cells, white blood cells, fibroblasts, pancreatic islet cells, cardiac and skeletal muscle cells, sperm, egg, and satellite cells.
  • tissue sources including, but not limited to, tumor cells, cells from the liver, kidney or central nervous system, epidermal keratinocytes, endothelial cells, stem cells, white blood cells, fibroblasts, pancreatic islet cells, cardiac and skeletal muscle cells, sperm, egg, and satellite cells.
  • Fetal, neonatal, juvenile and adult cells, tissues and organs all benefit from the protective aspects of the inventive gel-based carrier media.
  • cells from younger sources are highly sensitive to the mechanical stresses of preservation and transport, and thus are ideal candidates for the mechanically protective aspects of a gel-based preservation medium.
  • Cell lines and tissues from which cell lines are to be developed are suitable for storage and/or preservation in gel-based media.
  • organs and tissues from donor animals destined for transplantation into other animals, including humans have greater viability upon storage and transport in gel-based media.
  • Plant cells and tissues, such as those from transformed or transgenic sources may be stored and/or transported in gel-based media.
  • tissue grafts of plants are suitable samples for preservation and, if desired, transport in a gel-based medium.
  • Microbial and fungal cells would also benefit from storage and transport in a gel-based preservation medium.
  • microbial systems used in environmental remediation such as bacterially-mediated oil degradation or sewage treatment, may be successfully transported to a point of use in a gel-based preservation medium.
  • gelled preservation solutions permit maintenance of cells in suspension during periods of preservation. This maintenance of cellular suspension eliminates cell sedimentation and reduces mechanical stresses experienced by the cells subjected to storage and/or transport conditions.
  • the composition of the gelled preservation medium of the present invention is designed to provide a biochemical environment beneficial to cellular preservation. This protective environment is facilitated through the incorporation of an organ preservation solution as the principal diluent in which the gellation component of the medium is mixed.
  • Cellular monolayers are sheets of cells one to a few cell layers thick that are grown on an inert matrix in vitro. In the prior art, these monolayers are typically preserved in liquid based preservation media. Due to preservation in liquid-based media, monolayer separation from the growth matrix occurs during preservation. This separation results in the dissembling of the system and ultimately failure caused by the preservation process. Preservation of cellular monolayers using the gelled mediums of the present invention prevents cell-matrix separation during the preservation interval. Additionally, gelled media provide the same preservation benefits to cellular monolayers as is conferred upon cellular suspensions.
  • Biologic tissues are multi-layered cellular constructs that interact to perform a particular function.
  • the complexity of tissues ranges from engineered tissue constructs including a single cell type (i.e. EpidermTM, MatTech, Ashland, Mass., USA) to human skin grafts, to “micro-organs” (i.e. pancreatic islets), to whole human organs (i.e. kidneys, livers, etc.).
  • micro-organs i.e. pancreatic islets
  • whole human organs i.e. kidneys, livers, etc.
  • the gelled preservation media of the present invention enable preservation of tissue in a semi-solid preservation matrix, keeping the tissue structurally intact while affording the same protective benefits to the tissue as is conferred to individual cells and cell monolayers. Specifically, the reduction in the external mechanical shipping forces experienced by the tissues shipped in the inventive gel medium markedly improves tissue viability following preservation. For example, pancreatic islet cells preserved in a medium comprising gelatin and Hypothermosol® were afforded significant improvement of islet integrity and functionality following preservation (see FIG. 9 ).
  • Types of gellation material suitable for use in a gelled cell preservation medium include, but are not limited to, gelatin, carrageenan, agarose, collagen, laminin, fibronectin, plant-based gelling agents, and combinations thereof.
  • Plant-based gelling agents include gels produced from the roots, stems, tubers, fruit and seeds of plants.
  • the stock solution was mixed by swirling the container having the HTS and gelatin, although any suitable means of agitation may be employed.
  • the stock solution of gel-based medium was then warmed in a 40° C. water bath for 15-20 min with repeated swirling solution (once per minute) to dissolve the gelatin.
  • a 2% solution of gel-based medium was then prepared from this stock solution.
  • the desired volume depended upon the quantity of cells to be preserved. For example, for 100 ml of the 2% solution of gel-based medium, 14 ml of 14% stock solution of gel-based medium was combined with 86 ml of an HTS-Free Radical Scavenger (FRS) solution.
  • FFS HTS-Free Radical Scavenger
  • the FRS solutions discussed herein and shown in the figures include the anti-oxidant Trolox (vitamin E analogue) which is an anti-apoptotic agent. Aliquots of this 2% solution of gel-based medium were dispensed into 15 ml centrifuge tubes at 10 ml per tube, which were stored at 4° C. until used.
  • Trolox vitamin E analogue
  • Samples prepared in this manner may be stored for any desired time period at appropriate temperatures.
  • cells may be stored for less than 24 hours or for as long as about three days.
  • storage temperatures preferably range from about ⁇ 196° C. to about +30° C.
  • Temperatures are considered “normothermic” in a range between 31° C.-37° C. Temperatures are considered “hypothermic” in a range between 0° C. and 30° C. Cryopreservation generally occurs at temperature below 0° C., and may be achieved using a combination of a gel-based media with cryopreservants.
  • Sample tubes are removed from storage when the cells contained therein are to be used. Accordingly, the sample tubes in this instance were placed in a 37° C. water bath for 6-8 minutes to melt the gelled medium.
  • the gelling agent can be formulated into a preservation medium such that melting can occur at any desired temperature above or below 37° C. While samples were thawing, tubes were inverted every 30 seconds to maintain uniform temperature throughout the samples. Once the gelled medium containing sample cells melted, samples were immediately removed from the water bath. Cell samples were then gently centrifuged to form pellets, typically at 37° C. at 500 ⁇ g for 6 min. Cell sensitivity to heat stress will delimit tolerable temperature ranges for gel melting for a given biologic.
  • Centrifugation can be performed at room temperature, but the gel-based medium solution may partially resolidify at this temperature. Resolidification causes uncontrolled gel concentration within the cell pellet during centrifugation. For this reason centrifugation at temperatures between 30° C.-37° C. is preferred.
  • the gel-based medium supernatant was decanted from the cell pellet, which was then suspended in 12 ml of an appropriate cell culture medium at 37° C. to wash residual gel solution from the cells.
  • the samples were then gently centrifuged to pellet cells at 37° C., typically at 500 ⁇ g for 6 min.
  • the supernatant was decanted from the pelleted cells prior to resuspension to a desired cell density in an appropriate volume of cell culture media at 37° C. Suspended cells were then transferred to cell culturing vessels at the density desired for growth.
  • FIG. 1 shows post-storage recovery of MDCK (Madin Darby Canine Kidney) cells stored as monolayers in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 1 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • controls media, HTS, & HTS FRS
  • FIG. 1 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • the data in FIG. 1 shows the relative survival of MDCK cells following cold storage in either liquid (media (RPMI culture media), HypoThermosol (HTS), HypoThermosol-FRS (HTS-FRS) or gel-state preservation (HTS-gel; HTS-FRS gel) compared to pre-preservation cultured cells (control).
  • System viability was determined based on the metabolic function of each sample.
  • the data show that MDCK survival following 24 hours of gel-state preservation is equivalent to that of liquid storage demonstrating that gel-state storage is a viable and effective preservation modality.
  • the reduced viability at 1 hour post-storage is due to the depression of metabolism in the system following storage for which a recovery interval is necessary to resume proper function. This recovery period is what is responsible for the differential between the 1 hour and 24 hour data points.
  • the lower the redound differential the least change from 1 hour to day 1
  • FIG. 2 shows post-storage recovery of MDCK cells stored as monolayers in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 2 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • controls media, HTS, & HTS FRS
  • FIG. 2 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • the data in FIG. 2 shows the relative survival of MDCK cells following cold storage in either liquid or gel-state preservation compared to pre-preservation cultured cells (control). System viability was determined based on metabolic function of each sample. Overall, the data show that MDCK survival following 3 days of gel-state preservation is equivalent to that of liquid storage demonstrating that gel-state storage is a viable and effective preservation modality. The reduction of viability at 1 hour post-storage is due to the depression of metabolism in the system following storage for which a recovery interval is necessary to resume proper function. This recovery period is what is responsible for the differential between the 1 hour and 24 hour data points. Of note, the lower the redound differential (least change from 1 hour to day 1 ) the better preserved (less stressed) the cell system.
  • FIG. 3 shows post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 3 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • controls media, HTS, & HTS FRS
  • FIG. 3 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • media only media, hour data
  • HTS hour data
  • HTS hour data
  • HTSgel hour data
  • FIG. 4 shows post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 4 shows post-storage recovery at 1 hour and 1 day after the cells have been removed from the 4° C. storage conditions.
  • control media, HTS, & HTS FRS
  • FIG. 4 shows post-storage recovery at 1 hour and 1 day after the cells have been removed from the 4° C. storage conditions.
  • the data in FIG. 4 shows the relative survival of MDCK cells following cold storage in either liquid (media (RPMI culture media), HypoThermosol (HTS), HypoThermosol-FRS (HTS-FRS), or gel-state preservation (HTS-gel; HTS-FRS gel) compared to pre-preservation cultured cells (control).
  • System viability was determined based on the metabolic function of each sample.
  • the data from this series of studies demonstrates the advantage of gel state preservation in reducing the time dependent variability in samples during the recovery period. While overall sample viability is important, consistency during the recovery period following extended storage (such as 3 days) is also critical in cell suspension samples.
  • FIG. 5 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 1 day in maintenance and preservation solutions as gel formulations. More specifically, FIG. 5 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 22° C. storage conditions.
  • the data in FIG. 5 demonstrates the relative survival of islets following gel-state preservation at room temperature—a thermal range not possible for storage under other preservation regimes.
  • Islets were stored in three gel-based preservation media and compared to non-preserved cultured controls.
  • the data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day and 3 day data).
  • the dip in metabolic activity seen at 2 days post storage in all the stored samples was due to the metabolic cycling within the islet samples as the various cell populations within the islets began to re-synchronize following the preservation process.
  • Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations.
  • the xHTS-gel solution and the xHTS-FRS-gel solution shown in this figure differ in that the xHTS-FRS-gel solution includes the antioxidant Trolox; the two solutions are otherwise identical.
  • the HTS-gel solution shown in this figure is composed of approximately two times the concentrations of each of its components than the xHTS-gel solution. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 6 shows post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 3 days in maintenance and preservation solutions as gel formulations. More specifically, FIG. 6 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 22° C. storage conditions.
  • the data in FIG. 6 demonstrates the relative survival of islets following gel-state preservation at room temperature for 3 days—a thermal range not possible for storage under other preservation regimes. Islets were stored in three gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day and 3 day data). In contrast to FIG. 5 , no dip in metabolic activity is seen at 2 days post storage. This is due to the synchronization event of the islets occurring during the extended cold storage interval. Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 7 shows post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 8° C. for 1 day in maintenance and preservation solutions as gel formulations. More specifically, FIG. 7 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 8° C. storage conditions.
  • the data in FIG. 7 demonstrates the relative survival of islets following gel-state preservation at hypothermic temperatures for 1 day. Islets were stored in two low temperature gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day, 2 day, 3 day data). Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 8 shows post-storage recovery of human pancreatic Islets of Langerhans stored in suspension in culture plates at 8° C. for 3 days in maintenance and preservation solutions as gel formulations. More specifically, FIG. 8 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 8° C. storage conditions.
  • the data in FIG. 8 demonstrates the relative survival of islets following gel-state preservation at hypothermic temperatures for 3 days. Islets were stored in two low temperature gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day, 2 day and 3 day data). Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 9 a direct comparison of a media based approach, a HTS based approach and the approach of the present invention utilizing a Human Islet model for storage at 22° C. reveals that both the media and HTS based approaches fail to protect the islets to any degree. In contrast, the gel-based media facilitates substantial protection of the islets during storage, thereby resulting in increased viability post-storage.
  • the data in FIG. 9 demonstrates the comparison between islet survival following storage at 22° C. for 3 days in liquid or gel-state preservation.
  • the data are compared to typical islet survival following liquid storage in culture media (RPMI) for 1 day at 22° C. (industry standard shipment regime).
  • RPMI culture media
  • the data show that gel-state preservation and the control of molecular based cell death (using anti-apopotic agents, such as the ant-oxidant Trolox found in the Gelstor X-FRS) results in a significant improvement in cell survival compared to both short-term (Media 1 day) and long-term (media 3 day, HTS, and HTS-FRS) liquid storage.
  • metabolic activity was seen to be an 80% overall improvement at day 1 following 3 days of gel storage compared to 1 day liquid storage. This represents a significant advancement in preservation efficacy for islets going from one day to 3 days while providing for greater cell viability and function.
  • the present invention also includes a method of preserving a eukaryotic cell, tissue or organ by contacting the cell, tissue or organ with a hypothermic storage solution containing an agent that inhibits apoptotic cell death and a gelling agent, and storing the cell, tissue or organ and the hypothermic storage solution surrounding or perfusing the cell, tissue or organ.
  • the gel-state preservation is accomplished through use of a hypothermic storage solution that has a gelling agent. Reduction of the temperature of a sample in such a solution to below the gelling temperature results in the gel-state encapsulation of the cell, tissue or organ in that solution.
  • the inclusion of one or more anti-apoptotic agents aids in preventing the apoptotic cell death that normally occurs subsequent to this type of preservation.
  • Gel-state preservation involves the use of a hypothermic storage solution that forms a semi-solid matrix (gel), rather than a liquid or crystalline solid when exposed to low temperatures, specifically, temperatures below the gelling point of the solution.
  • Gel-state preservation as the term is used herein involves the non-crystalline liquid to semi-solid phase transition of the surrounding or perfused hypothermic storage solution thereby encapsulating the cell, tissue, or organ.
  • This gel-state thus does not involve the formation of macromolecular crystals (such as ice crystals) within or outside of cells, tissues or organs placed into or perfused with such a solution. This is in contrast to cryopreservation, in which ice crystals are permitted to form outside of such cells, or hypothermic storage where cells are bathed in a liquid media for the preservation interval.
  • gel-state preservation involves the steps of contacting the cells, tissue or organ with the hypothermic storage solution that contains one or more agents that inhibit apoptosis, and then reducing the temperature below the gelling point (solidification point) of the solution, such that a gel-like semi-solid is formed.
  • the rate of cooling depends upon the solution, but is generally in the range of 0.5° C. to 50° C. per minute, most often approximately 1° C. to 20° C. per minute, e.g., approximately 1° C. per minute, approximately 5° C. per minute, approximately 10° C. per minute, approximately 15° C. per minute or approximately 20° C. per minute.
  • the skilled artisan can identify an optimal rate of cooling for a given combination of the gel-state solution and tissue.
  • Gel-state formation may be monitored in several ways.
  • One of the simplest ways is to observe the solution (solid vs. liquid above 0° C.) or by observing or measuring the transmission of light through the gel.
  • the gel-state sample is then stored below the gelling point.
  • the gelling point generally ranges from +25° C. to ⁇ 196° C. (liquid nitrogen temperature). Further reduction in temperature below the gelling point is often desirable. Such preservation maintains the semi-solid state of the gel without permitting melting or crystallization within and outside of cells.
  • the step of contacting cells, tissues or organs with a hypothermic storage solution differs depending upon whether the sample is cells, a tissue or an organ.
  • cells or relatively thin (e.g., up to about 2-5 mm) or porous tissues (e.g., skin or artificial skin) can simply be immersed in the hypothermic storage solution prior to chilling to cause gelling.
  • This immersion is preferably performed at or near 22° C. or 4° C.
  • the length of time necessary for immersion will clearly vary with the thickness of the tissue, with longer times necessary as the tissue becomes thicker.
  • One of skill in the art can readily evaluate and adjust the time necessary for the immersion of a given sample or type of sample by evaluating the survival, recovery and degree of apoptosis occurring in samples immersed for varying lengths of time before the initiation of gelling.
  • organs e.g., kidney, liver, heart, lung, etc.
  • Methods for organ perfusion vary for different organs but are well known in the art.
  • an important parameter is the viscosity of the solution. More porous organ tissues (e.g., liver) may be perfused with relatively higher viscosity solutions than can less porous tissues (e.g., heart). Because of differences in the ability to perfuse different tissues, it may be necessary in some cases to perfuse with an increasing gradient, or with stepwise increases, of the concentration of hypothermic storage solution until a concentration is achieved such that the tissue will gel throughout upon subsequent reduction of temperature.
  • apoptotic pathway The precise cellular mechanisms regulating apoptosis are not completely known. However, various aspects of the apoptotic pathway have been elucidated. For example, alteration of the ionic environment may be necessary to activate or inhibit the endonucleases relevant to the process of apoptotic nuclear degradation (e.g., physiologic concentrations of Zn++ are known to inhibit DNA fragmentation and apoptosis). Treatment of certain cells with inhibitors of macromolecular synthesis, such as Actinomycin D to block RNA synthesis or cyclohexamide to block protein synthesis, induces apoptosis.
  • inhibitors of macromolecular synthesis such as Actinomycin D to block RNA synthesis or cyclohexamide to block protein synthesis
  • Completion of the apoptotic process appears to depend upon the regulated expression of various gene products associated with the promotion or suppression of gene activated cell death, particularly gene products involved with cell cycle regulation.
  • overexpression of the cell-death inhibiting agents Bcl-2 and Bcl-xL prevents the release of cytochrome C.
  • Cytochrome C is thought to activate the caspases, a group of proteases known for cleaving substrates responsible for the changes associated with apoptosis.
  • Enhanced levels of Bax, a pro-apoptotic member of the Bcl-2 family promote cytochrome C release and subsequent apoptosis of cells.
  • c-myc, c-jun and c-fos may promote either cell growth or cell death, depending upon the circumstances surrounding their expression.
  • Another trigger for apoptosis involves oxidative stress.
  • antioxidants and free-radical scavengers have been demonstrated to inhibit the initiation of apoptosis.
  • programmed cell death involves an intricate cascade of cellular events.
  • hypothermic storage is among the numerous triggers for apoptosis.
  • the gel-state preservation of cells tends to cause a portion of the cells to undergo apoptosis and/or necrosis when they are removed from gel-state storage.
  • the inclusion, in the hypothermic storage solution used for gel-state preservation, of one or more agents that alone or collectively inhibit apoptosis can increase the survival and recovery of the cells.
  • Agents useful according to the invention can function at any stage of the apoptotic pathway, e.g., by modulating the function or expression of one or more nuclear or cytoplasmic polypeptide mediators of the pathway (e.g., gene products in the regulatory cascade such as Bcl-xL, Bcl-2 or Bax, cytochrome C, caspase enzymes, etc.) or by avoiding or countering the effects of oxidative stress known to trigger apoptosis (e.g., through use of antioxidants, free radical scavengers or agents that modulate the function or expression of nitrous oxide synthase).
  • nuclear or cytoplasmic polypeptide mediators of the pathway e.g., gene products in the regulatory cascade such as Bcl-xL, Bcl-2 or Bax, cytochrome C, caspase enzymes, etc.
  • oxidative stress e.g., through use of antioxidants, free radical scavengers or agents that modulate the function or expression
  • One useful way to categorize inhibitors of apoptosis is to consider those that interact with a cellular polypeptide factor that participates in the generation, propagation or execution of an apoptotic signal to be one category, and those that inhibit by other means as a second category.
  • the interaction of the agent with the polypeptide factor is a physical, i.e., binding, interaction.
  • an agent that physically interacts with and inhibits the function of a caspase enzyme would fall into this category.
  • the agent need not necessarily interact with or bind a polypeptide, but rather acts by, for example scavenging free radicals or maintaining the redox status quo.
  • agents in the second category include, for example, Vitamin E and other agents listed in Table II.
  • a composition that inhibits apoptosis includes at least one agent that reduces the extent of apoptotic cell death, but may include, for example, two or more such agents, where each such agent preferably, but not necessarily, interacts with a different part of an apoptotic pathway.
  • the composition includes two or more agents that together have an inhibitory effect on apoptosis that neither has on its own, or that together have a synergistic effect.
  • An anti-apoptotic agent useful according to the invention will inhibit apoptosis, as measured by a TUNEL assay, by at least 10% relative to a sample subjected to the same apoptotic induction conditions without the addition of the anti-apoptotic agent.
  • cellular polypeptide targets involved in the promotion of apoptosis are listed in Table IA.
  • an anti-apoptotic agent useful according to the invention can act on one or more of these cellular polypeptide targets.
  • Table IB also lists exemplary targets involved in the prevention of apoptosis.
  • an anti-apoptotic agent can act to increase or stabilize the activity of one or more of these cellular polypeptide targets.
  • caspase inhibitors As noted, an important class of anti-apoptotic agents is the caspase inhibitors, a number of which are known and commercially available.
  • One class of caspase inhibitors comprises fluoromethylketone (FMK) derivatives of peptides that mimic the recognition and cleavage sites for the target caspase enzymes.
  • FMK fluoromethylketone
  • caspase-1 inhibitor YVAD-FMK (benzyloxycarbonyl tyrosylvalylalanyl aspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 1
  • caspase-3 inhibitor DEVD-FMK (benzyloxyycarbonyl aspartyl glutamylvalylaspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 3 but also inhibits caspase 7, caspase 10, and caspase 6 in decreasing order of binding affinity
  • caspase-6 inhibitor VEID-FMK (benzyloxycarbonyl valylglutamyl isoleucylaspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 6 and also inhibits caspases 3, 7, and 8 in decreasing order of binding affinity
  • caspase-8 inhibitor LETD-FMK (benzyloxycarbonyl leucylglutamylthreonyla
  • FMK-peptide derivatives useful for the inhibition of caspases include, but are not limited to caspase-2 inhibitor VDVAD-FMK, caspase-4 inhibitor Z-LEVD-FMK, caspase-5 inhibitor WEHD-FMK, caspase-10 inhibitor AEVD-FMK and caspase-13 inhibitor LEED-FMK.
  • caspase inhibitors, and cocktail mixtures of them are commercially available from, for example, Gentaur Molecular Products (Brussels, Belgium).
  • Additional anti-apoptotic agents include the calpain inhibitors (e.g., Calpain Inhibitor Z-LLT-FMK) and the cathepsin B&L inhibitor Z-Phe-Phe-FMK. These inhibitors are also available from Gentaur Molecular Products. Others include Calpain Inhibitor I (N-Acetyl-Leu-Leu-norleucinal, available from Roche Diagnostics), Calpain Inhibitor II (N-Acetyl-Leu-Leu-methioninal, available from Roche Diagnostics), and CTX295 (a dipeptide alpha-ketoamide compound which inhibits calpain (DiBiasi et al., 2001, J. Virol. 351-361)). Additional anti-apoptotic agents useful according to the invention are listed in Table II.
  • the amount of an anti-apoptotic agent or agents useful in a gel-based storage solution according to the invention will vary depending on the nature of the inhibitor and its target, if the target is known (there is no requirement that the specific target of the apoptotic inhibitor useful according to the invention be known). Generally, the amount of anti-apoptotic agent useful will be that amount or concentration determined by the user to provide at least 10% (and preferably greater) inhibition in apoptosis following removal from the gelled state relative to a sample that was in the gelled state in the absence of the inhibitor or inhibitors. The concentration of anti-apoptotic agent useful according to the invention will further not be associated with significant (i.e., greater than 2% cell death over the course of treatment or contact) toxic effects.
  • Bcl-2 Mitochondrial Anti- Initiation, apoptotic protein
  • Execution Bcl-x Mitochondrial Anti- Initiation, apoptotic protein
  • Execution IAP Inhibitor of Apoptosis Initiation, Protein
  • Execution RAS Receptor mediated pro- Initiation survival signal AKT Anti-apoptosis signal Initiation, Execution TRAF2 (TNF Receptor Associated Initiation Factor 2)
  • Flavonoids Vitamin E Vitamin C Vitamin D Beta Carotene (Vitamin A) Pycnogenol Super Oxidedismutase N-Acetyl Cysteine Selenium Catechins Alpha Lipoic Acid Melatonin Glutathione Zinc Chelators Calcium Chelators L-Arginine
  • apoptosis There are a number of ways for one to measure the extent of apoptosis occurring following the gel state preservation of a sample containing cells. As noted, one of the hallmarks of apoptosis is a regular DNA fragmentation pattern leading to a “ladder” when genomic DNA is subjected to gel electrophoresis. This assay is well known in the art, and can be performed as described in U.S. Pat. No. 6,045,990.
  • TUNEL assay Terminal deoxynucleotidyl transferase mediated dUTP nick end labeling
  • This assay measures the enzymatic incorporation of labeled dUTP at the nick or breaks in DNA that accompany apoptosis.
  • the assay is well known to those of skill in the art, and kits for performing the assay are available, for example, from Intergen (ApopTagTM kit, Intergen Company, Purchase, N.Y.).
  • Kits for this are also commercially available, e.g., from Promega (Madison, Wis.).
  • Annexin V binding to cells can be measured.
  • One of the early events of apoptosis is the loss of membrane asymmetry of phospholipids.
  • Phosphotidylserine normally located in the inner leaflet of the membrane, redistributes and appears in the outer leaflet at the early stage of apoptosis.
  • Annexin V binds specifically to phosphotidylserine on apoptotic cell surfaces in the presence of calcium, and can be used as a marker for apoptosis. Binding can be measured by monitoring a fluorescent tag on the Annexin V, e.g., fluorescein.
  • the process for the removal of cells, tissues or organs from the gel-state state will vary depending upon the cell, tissue or organ preserved.
  • the general process involves warming the sample above the gelling point, followed by rinsing or dilution of the sample to remove the hypothermic storage solution, followed by re-establishment in culture or by implantation into a recipient.
  • Rinsing can be performed by simple sample dilution (e.g. for isolated cells and cell suspensions), immersion (e.g., for cells or relatively thin tissues) or by perfusion (e.g., for organs).
  • immersion e.g., for cells or relatively thin tissues
  • perfusion e.g., for organs
  • an anti-apoptotic composition can advantageously be added to any such rinse solution.
  • gel-state storage encompasses storage for a matter of hours or for a matter of weeks, months or years.
  • the level of cell survival acceptable for such storage will depend upon the cell, tissue or organ type stored and the reason for its storage (e.g., transplantation versus research uses), but will advantageously be higher than is practicable using prior art technologies.
  • a cell, tissue or organ is used for human transplantation, it is advantageous to maximize the survival and ensuing function of the cell, tissue or organ.
  • gel-state storage is considered successful if it is accompanied by death of about 20% or fewer of the cells, preferably 10% or less, and most preferably less than 5%, up to and including full viability and function.
  • the storage efficacy of numerous cell preservation solutions can be assessed using a number of assays.
  • assays include enzyme synthesis (Pahernik et al., 1996, Cryobiology 33: 552), potassium content (Fisher et al., 1996, Cryobiology 33: 163), trypan blue exclusion (Rodriguez et al., 1995, Cell Transplant. 4: 245), neuronal outgrowth and myelination (Levi et al., 1994, Glia 10: 121), contraction (Lopukhin et al., 1996, Cryobiology 33: 178), ATP content (Zhang et al., 1996, J. Surg. Res.
  • a preferred assay for viability uses the alamarBlue® non-toxic dye.
  • the assay using alamarBlue® dye measures viability by the metabolic conversion of the agent to a fluorescent form detectable with standard fluorescence detection equipment. The assay does not indicate whether or not test cells are functioning in a tissue specific manner.
  • one of the key attributes of the alamarBlue® dye assay that is not shared by most of the other viability or function assays described previously is the ability of alamarBlue® dye to be used repetitively, day after day, as a non-toxic indicator. This has been shown to be critically important to some cold-stored tissues such as human skin cells.
  • alamarBlue® dye viability testing following gel-state storage is as follows.
  • Cells e.g., cultured cells or cells isolated from a patient
  • isotonic buffer solution e.g., phosphate buffered saline, Hanks' Balanced Salt Solution (HBSS), etc.
  • HBSS Hanks' Balanced Salt Solution
  • the cell sample is cooled and maintained for a given time period at low temperature, e.g., 4° C., after which the sample is warmed such that the solution returns to the liquid state.
  • the gel-state storage solution is removed, cells are washed in isotonic buffer solution or cell culture medium and plated for growth in appropriate cell culture medium. After a given period of time (generally about 24 hours, but advantageously later, e.g., 48 hours or more) medium is removed from the recovered cultures, and a 1:20 dilution of alamarBlue® dye (Accumed International, Westlake, Ohio) in HBSS without phenol red is placed on the cells.
  • Fluorescence of the converted dye is measured using, for example, a CytoFluor 2350 (Millipore Corporation, Bedford, Mass.) or a CytoFluor II (PerSeptive Biosystems, Inc., Cambridge, Mass.) apparatus with a 530 nm excitation/590 nm emission filter set. Following the assay, cells can be provided with fresh medium and re-cultured with repeat assays at later times as desired.
  • Calcein-AM Molecular Probes, Eugene, Ore.
  • Calcein-AM is suspended at 1 mg/ml in DMSO and then mixed with HBSS at 1:100. This solution is then contacted with recovered cells for one hour, followed by washing with HBSS. Fluorescence of the retained dye, indicative of dead cells, is then measured using a 485 nm excitation/530 nm emission filter set.
  • the present invention preserves various organs, tissues and cells using semi-solid gel-state storage.
  • Organs including but not limited to lung, liver, heart, kidney, gut, eye and skin can be preserved according to the invention prior to transplantation in a recipient patient.
  • Tissues such as bone marrow and cells such as erythrocytes and leukocytes can be preserved for long term storage according to the invention.
  • tissues for forensic and pathology records may be preserved without significant loss of viability.
  • Cell lines for therapeutic and research interests can be preserved for long periods by applying the invention. It is contemplated that variations of the invention can be applied for long term preservation of entire multicellular organisms.

Abstract

Gel-based medium compositions and a method of use thereof in normothermic, hypothermic or cryopreservative storage and transport of cell samples are described. These gel-based compositions preferably include an agent that inhibits apoptosis, together with a gelling agent. Such gel-based medium compositions protect various cell samples, such as animal or plant organs, tissues and cells, from the mechanical, physiological and biochemical stresses inherently associated with liquid preservation techniques.

Description

    REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation in part application of co-pending application Ser. No. 10/166,732, filed Jun. 12, 2002, entitled “NORMOTHERMIC, HYPOTHERMIC AND CRYOPRESERVATION MAINTENANCE AND STORAGE OF CELLS, TISSUES AND ORGANS IN GEL-BASED MEDIA”, which is a divisional patent application of application Ser. No. 09/757,694, filed Jan. 11, 2001, entitled “NORMOTHERMIC, HYPOTHERMIC AND CRYOPRESERVATION MAINTENANCE AND STORAGE OF CELLS, TISSUES AND ORGANS IN GEL-BASED MEDIA”, now U.S. Pat. No. 6,632,666, which claims an invention which was disclosed in Provisional Application No. 60/176,009, filed Jan. 14, 2000, entitled “NORMOTHERMIC, HYPOTHERMIC AND CRYOPRESERVATION MAINTENANCE AND STORAGE OF CELLS, TISSUES AND ORGANS IN GEL-BASED MEDIA”. The benefit under 35 USC §119(e) of the United States provisional application is hereby claimed, and the aforementioned applications and patents are hereby incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The invention relates to the gel state preservation of cells, tissues and organs. In particular, the invention relates to the hypothermic preservation or storage of cells, tissues or organs in the semi-solid, gel-like state.
  • 2. Description of Related Art
  • Today, limited normothermic, hypothermic and cryopreservative maintenance and storage of plant or mammalian cells, tissues and organs (biologics) is accomplished in liquid media. Success is limited, in part, due to damage that occurs during shipment (transport), most often associated with mechanical trauma.
  • Preservation and transport (herein referred to as preservation) of biologics (cells, tissues, and organs) has traditionally been achieved through “suspension” in a liquid preservation medium. These media include, but are not limited to a simple saline solution, cell culture media, and preservation solutions such as University of Wisconsin (UW) solution (Viaspan® solution), Euro-Collins™ solution, and Hypothermosol® solution (Bio Life Solutions, Inc., Owego, N.Y., USA). Inherent in this liquid preservation approach is that the liquid environment confers no physical support network for the biologic during preservation and transport. Due to this lack of physical support upon preservation, biologics are exposed to numerous physical stresses during storage and shipment. These stresses include, but are not limited to, sedimentation, mechanical “jarring”, compaction in a liquid column, shaking, vibration, shearing forces, ice damage, and the like. As a result of these mechanical stresses plus additional biochemical stresses inherently associated with biologic preservation in liquid, a significant level of cellular death is initiated during and following the preservation interval. Consequently, failure of the biologic ensues due to this preservation-initiated cell death.
  • The preservation of eukaryotic cells, tissues or organs is commonly carried out by chilling them sufficiently to slow or halt metabolic processes that require ongoing support by an organism or the environment to maintain viability. The preservation of cells, tissues or organs by such chilling is generally referred to as hypothermic preservation or hypothermic storage.
  • There are two broad types of hypothermic storage for cells, tissues or organs. The first involves storage at temperatures above the freezing point of the solution or medium in which the sample is suspended or immersed or with which the sample is perfused. For example, such temperatures may be in the range of about 10° C. to 0° C. Such conditions are appropriate only for short term storage, generally on the order of hours to several days to about a week.
  • The second broad type involves storage at lower temperatures, for example, as low as −80° C. to −196° C. Storage under these conditions is more appropriate for longer periods of time. In this second broad category, there are two general sub-classes of preservation approach. The first sub-class involves the freezing of the samples in a medium or solution that permits the formation of ice crystals. For this approach, cryoprotective agents (e.g., dimethyl sulfoxide (DMSO), glycerol) are added that mitigate the effect of the formation of ice crystals, essentially by causing dehydration of the cells that prevents intracellular water crystal formation. This approach is referred to herein as “cryopreservation.”
  • The basic challenge of hypothermic storage is to preserve the material in a state that can be reversed without causing extensive cell damage or cell death. Approaches (solutions and methods) that minimize the accumulation of sub-lethal damages such as oncotic balance, energy deprivation, cellular waste accumulation, ionic balance, as well as formation of ice crystals in or around cells are well known to aid in the survival and ultimate recovery of material stored at hypothermic temperatures. However, even when these factors are controlled, as in the case of hypothermic storage and cryopreservation, there remains an associated degree of cell death. Cell death is known to occur by two different mechanisms. The first, necrotic cell death or necrosis, is not mediated by a specific cellular pathway. Necrosis is characterized by the loss of cell membrane integrity resulting in cell swelling, and is caused by a number of pathological agents. DNA in cells that undergo necrosis is cleaved in a random fashion. Thus, the DNA from cells that have undergone necrosis appears as a continuous smear when subjected to gel electrophoresis. The second cell death mechanism, apoptosis or programmed cell death, is the result of the activation of a specific biochemical pathway involving a cascade of biochemical activation steps that ultimately result in the death of the cell. Apoptosis is characterized by cell shrinkage, intact plasma membranes, and non-random cleavage of DNA at an approximately 180 nucleotide interval, evidenced by a ladder of DNA cleavage products upon gel electrophoresis of genomic DNA. Apoptosis is reviewed, for example, by Kerr et al., 1994, Cancer 73: 2013 and Evan & Littlewood, 1998, Science 281: 1317.
  • Cell death accompanying hypothermic storage involves an apoptotic component. U.S. Pat. No. 6,045,990, incorporated herein by reference, demonstrates, in part, that survival and recovery from cryopreservation can be enhanced by the inclusion of anti-apoptotic agents in the preservation medium.
  • U.S. Pat. No. 6,921,633 discloses methods for the hypothermic preservation of cells, tissues and organs in the vitreous state. This preservation can only be accomplished at very low temperatures. Preservation of cells, tissues, and organs in a vitreous state requires the utilization of high concentrations of toxic agents coupled with controlled reduction of sample temperature to ultra-low temperatures (<−140° C.) to cause the formation of a glass (amorphous solid). The extent of cell survival following vitreous preservations can be significantly improved by the inclusion of anti-apoptotic agents into the preservation medium. This patent is incorporated herein by reference.
  • There is a need in the art for improved methods of hypothermic preservation over a wide range of temperatures.
  • SUMMARY OF THE INVENTION
  • The present invention includes methods and compositions for the preservation of cells, tissues or organs in the semi-solid state.
  • The invention includes gel-based medium compositions for normothermic, hypothermic or cryopreservative transport and/or storage of plant tissues or cells and animal organs, tissues or cells. In one embodiment, the gel-based compositions include a cell maintenance and preservation medium and a gelling agent. In particular, mammalian samples, such as human and animal organs, tissues and cells, may be preserved in the gel-based media compositions. In a preferred embodiment, the cell maintenance and preservation medium is liquid.
  • In one embodiment, a method of preserving a eukaryotic cell, tissue or organ includes the step of contacting the cell, tissue or organ with a semi-solid, gel-like storage solution. The storage solution preferably includes a composition that inhibits the activation and progression of molecular-based cell death cascades (apoptosis and necrosis) and a concentration of a semi-solid gelling agent that is sufficient for gelling of the solution. The storage solution also preferably includes agents which protect cells from ice-related damage (including but not limited to dimethyl sulfoxide, glycerol, and propanediol). The method also includes the step of preserving the cell, tissue or organ, where the cell, tissue or organ becomes encapsulated within the semi-solid gel-state preservation matrix.
  • The molecular based cell death control during gel-state preservation is preferably accomplished through use of a hypothermic storage solution which contains a gelling agent and agents designed to inhibit the activation and progression of apoptotic and necrotic cell death cascades. The inclusion of one or more anti-apoptotic agents aids in preventing the apoptotic cell death that normally occurs subsequent to this type of preservation.
  • In one embodiment, the gel-state preservation is performed at a temperature of +37° C. to −0° C.
  • In another embodiment, the gel-state preservation is performed at a temperature of −1° C. to −196° C., and more preferably at a temperature of −80° C. to −196° C.
  • In another embodiment, the composition that inhibits apoptosis includes an agent that interacts with a polypeptide that participates in an apoptotic pathway. In one embodiment, the agent inhibits the activity of the polypeptide. In another embodiment, the agent maintains or potentiates the activity of the polypeptide.
  • In another embodiment, the agent is selected from the group consisting of a caspase inhibitor, a calpain inhibitor, and an inhibitor of nitrous oxide synthase. In another embodiment, the composition that inhibits apoptosis includes an antioxidant. In another embodiment, the composition that inhibits apoptosis includes an agent selected from the group consisting of a free radical scavenger, a zinc chelator, and a calcium chelator.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 graphically depicts post-storage recovery of MDCK (Madin Darby Canine Kidney) cells stored as monolayers in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 2 graphically depicts post-storage recovery of MDCK cells stored as monolayers in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 3 graphically depicts post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 4 graphically depicts post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel).
  • FIG. 5 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 1 day in maintenance and preservation solutions as gel formulations.
  • FIG. 6 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 3 days in maintenance and preservation solutions as gel formulations.
  • FIG. 7 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 8° C. for 1 day in maintenance and preservation solutions as gel formulations.
  • FIG. 8 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans stored in suspension in culture plates at 8° C. for 3 days in maintenance and preservation solutions as gel formulations.
  • FIG. 9 shows post-storage function of human islets stored for 3 days at 22° C.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention preserves cells in the gel-state (semi-solid matrix). Gel-state preservation offers a number of advantages over liquidous preservation where samples can be suspended in a liquid which then solidifies, placing the specimen into a semi-solid suspended state for preservation. Gel state preservation provides for a number of additional protective avenues beyond liquidous storage including, but not limited to, physical protection from sheer stress and strain, mechanical damage, specimen settling and clumping. In the area of cryopreservation, gel-state preservation offers the benefit of altering the formation and structure of ice crystals thereby reducing the negative physical effects of sub-freezing specimen storage.
  • As used herein, the term “gel-state” means establishing a semi-solid matrix state in a solution and in cells, tissue or organs suspended in or perfused with that solution. A “gel-state” is a semi-solid formed from a liquid without the formation of crystals. A gel-state refers more particularly to a solid formed from a liquid without the formation of ice crystals. Gel-state preservation is accomplished by reducing the temperature of a solution below the gel solidification point, a variable temperature point based on specific composition ranging from 37° C. to −196° C., such that the cells, tissue or organs are suspended in or perfused with that solution. That is, “gel-state,” as it is used herein, is when a cell, tissue or organ is encapsulated in a semisolid matrix (i.e., in the hypothermic storage solution).
  • “Hypothermic storage solution” refers to a solution in which cells, tissues, or organs can be stored at temperatures below physiological temperature. Preferred hypothermic storage solutions are described below.
  • As used herein, a “composition” can have one or more component elements.
  • As used herein, the term “inhibit” means to reduce an activity by at least 10%, and preferably more, e.g., 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more, up to and including 100% relative to that activity that is not subject to such inhibition. Thus, an agent that inhibits apoptosis inhibits apoptosis and/or necrosis by at least 10% relative to a sample subject to the same apoptotic stimulus but absent the agent.
  • As used herein, the term “enhance” means to increase an activity by at least 10%, and preferably more, e.g., 20%, 50%, 75% or even 100% or more (e.g., 2×, 5×, 10×, etc.) relative to that activity that is not subject to such enhancement.
  • As used herein, an “anti-apoptotic agent” refers to a composition that inhibits apoptosis and/or necrosis triggered by or accompanying gel-state preservation of a cell, tissue or organ. Anti-apoptotic agents fall into two broad classes, those that interact with a polypeptide that participates in an apoptotic pathway (e.g., participation in the cellular generation, propagation or execution of an apoptotic signal) and those that inhibit by other means, for example, frequently by avoiding or countering the effects of oxidative stress that tend to activate the apoptotic program. In the first broad class of agents, which target polypeptide factors, there are two sub-categories: a) agents that inhibit the activity of polypeptides that participate in the pathway, and b) agents that maintain or potentiate the activity of polypeptides whose normal role it is to prevent apoptosis. In one embodiment, the present invention uses an anti-apoptotic agent or composition that only targets one or more polypeptide targets. In another embodiment, the present invention uses an anti-apoptotic agent or composition that only targets other aspects of the apoptotic process, e.g., changes in redox status or potential. In another embodiment, the present invention uses one or more agents or compositions that act upon either of these sites.
  • As used herein, the term “maintains or potentiates the activity of a polypeptide” refers to compositions or agents that prevent a decrease or cause an increase in a given activity of a given polypeptide. For example, when a polypeptide is active in preventing apoptosis, an agent that maintains or potentiates the activity of that polypeptide prevents a decrease in the anti-apoptotic activity of that polypeptide, thereby preventing or inhibiting apoptosis (i.e., inhibiting apoptosis by at least 10% relative to a sample not treated with that agent and subject to the same apoptotic stimulus).
  • As used herein, the term “composition that interacts with a polypeptide that participates in an apoptotic pathway” refers to a composition comprising one or more agents that, alone or together, physically interact with a polypeptide target in the apoptotic pathway such that apoptosis is decreased or inhibited. Non-limiting examples of such polypeptides are listed in Table I A and B.
  • As used herein, the term “polypeptide that participates in an apoptotic pathway” refers to a polypeptide, at least one function of which is to promote or inhibit apoptosis. Thus, one class of such polypeptides includes a polypeptide that acts to promote apoptosis—the inhibition of its function or expression, e.g., a specific protease activity, leads to an inhibition of apoptosis. Another class of such polypeptides includes a polypeptide that acts to inhibit apoptosis—the activation or maintenance of its function or expression, e.g., Bcl-2 activity, also leads to an inhibition of apoptosis.
  • Hypothermic storage solutions useful for semi-solid or gel-state preservation, known and commercially available in the art, can be readily adapted for use in the methods and compositions of the present invention. These include, but are not limited to, various tissue culture medias, saline solutions, HypoThermosol® solution, Viaspan® solution, CryoStor™ solution, Celsior® solution, HTK® solution, Euro-collins™ solution, Normosol® solution, Cardisol™ solution, Unisol® solution, and Plasmalite® solution.
  • Gel-Based Preservation Solutions
  • To reduce and even eliminate damage associated with mechanical factors arising from cell storage and transport, gel-based maintenance and preservation media have been developed to sustain biologics in supporting environments of various viscosity. The gel-based media are rigid or highly viscous at temperatures equal to or lower than 28° C. but liquid at 37° C. Liquid maintenance and preservation media (i.e., Hypothermosol® solution, Viaspan® solution, also called University of Wisconsin solution, EuroCollins™ solution, Cardisol™ solution, Unisol® solutions, tissue culture media, etc.) are converted to a gel of appropriate viscosity necessary to match cell, tissue or organ type with either animal or plant derived gelling agents (1-2% by volume). Suitable combinations of such liquid maintenance and preservation solutions may be used if desired.
  • A preferred organ preservation solution is Hypothermosol® solution. Hypothermosol® solution is composed of (a) one or more electrolytes selected from the group consisting of potassium ions at a concentration ranging from about 10-145 mM, sodium ions ranging from about 10-120 mM, magnesium ions ranging from about 0.1-10 mM, and calcium ions ranging from about 0.01-1.0 mM, (b) a macromolecular oncotic agent having a size sufficiently large to limit escape from the circulation system and effective to maintain oncotic pressure equivalent to that of blood plasma and selected from the group consisting of human serum albumin, polysaccharide and colloidal starch, (c) a biological pH buffer effective under physiological and hypothermic conditions, (d) a nutritive effective amount of at least one simple sugar, (e) an impermeant and hydroxyl radical scavenging effective amount of mannitol, (f) an impermeant anion impermeable to cell membranes and effective to counteract cell swelling during cold exposure, the impermeant ion being at least one member selected from the group consisting of lactobionate, gluconate, citrate and glycerophosphate-like compounds, (g) a substrate effective for the regeneration of ATP, the substrate being at least one member selected from the group consisting of adenosine, fructose, ribose and adenine, and (h) at least one agent which regulates cellular levels of free radicals.
  • Adjusting the formulation of 1) the carrier medium, in particular, a cell maintenance and preservation solution, 2) gel-type and 3) gel concentration produces an appropriate long-term (up to two weeks) shipping environment, which is commercially known as GELSTOR® solution (Biolife Solutions, Inc., Owego, N.Y., USA). Procedurally, a biologic of choice is exposed to liquefied gel at 37° C., and thereafter cooled to a desired preservation temperature for maintenance and shipment. Upon arrival or point-of-use, the gel is warmed to 37° C. whereupon it liquefies, and may be decanted. The biologic may then be rinsed with an appropriate medium and is ready for use.
  • The gel-based preservation and maintenance technology disclosed herein provides a semi-solid preservation matrix that facilitates protection of various cells from mechanical stresses associated with biologic preservation and transport. In addition to physical protection, gel-based preservation media provide physiological and biochemical protection during the storage and transport of cells, organs and tissues.
  • A variety of animal and plant cells may be stored and transported in gel-based media, including human cells and animals cells from numerous tissue sources, including, but not limited to, tumor cells, cells from the liver, kidney or central nervous system, epidermal keratinocytes, endothelial cells, stem cells, white blood cells, fibroblasts, pancreatic islet cells, cardiac and skeletal muscle cells, sperm, egg, and satellite cells. Fetal, neonatal, juvenile and adult cells, tissues and organs all benefit from the protective aspects of the inventive gel-based carrier media. In particular, cells from younger sources are highly sensitive to the mechanical stresses of preservation and transport, and thus are ideal candidates for the mechanically protective aspects of a gel-based preservation medium. Conversely, cells from older sources are highly susceptible to damage from oxidative stresses incurred during preservation and transport, meaning these cells particularly benefit from the physiological and biochemical protection afforded by gel-based preservation media.
  • Cell lines and tissues from which cell lines are to be developed are suitable for storage and/or preservation in gel-based media. Likewise, organs and tissues from donor animals destined for transplantation into other animals, including humans, have greater viability upon storage and transport in gel-based media. Plant cells and tissues, such as those from transformed or transgenic sources may be stored and/or transported in gel-based media. Likewise, tissue grafts of plants are suitable samples for preservation and, if desired, transport in a gel-based medium. Microbial and fungal cells would also benefit from storage and transport in a gel-based preservation medium. For example, microbial systems used in environmental remediation, such as bacterially-mediated oil degradation or sewage treatment, may be successfully transported to a point of use in a gel-based preservation medium.
  • Traditionally, preservation and transport of single cells has relied on suspension of cells in a liquid-based medium. Such liquid-based preservation regimes result in the settling of cells to the bottom of the preservation container. This settling causes an inhomogeneous distribution of the cells in the medium resulting in inefficient exposure of the cells to the preservation solution, as well as the exposure of the cells to mechanical stresses of “jarring and shearing” experienced during shipment.
  • In contrast, gelled preservation solutions permit maintenance of cells in suspension during periods of preservation. This maintenance of cellular suspension eliminates cell sedimentation and reduces mechanical stresses experienced by the cells subjected to storage and/or transport conditions. Additionally, the composition of the gelled preservation medium of the present invention is designed to provide a biochemical environment beneficial to cellular preservation. This protective environment is facilitated through the incorporation of an organ preservation solution as the principal diluent in which the gellation component of the medium is mixed.
  • Cellular monolayers are sheets of cells one to a few cell layers thick that are grown on an inert matrix in vitro. In the prior art, these monolayers are typically preserved in liquid based preservation media. Due to preservation in liquid-based media, monolayer separation from the growth matrix occurs during preservation. This separation results in the dissembling of the system and ultimately failure caused by the preservation process. Preservation of cellular monolayers using the gelled mediums of the present invention prevents cell-matrix separation during the preservation interval. Additionally, gelled media provide the same preservation benefits to cellular monolayers as is conferred upon cellular suspensions.
  • Biologic tissues are multi-layered cellular constructs that interact to perform a particular function. The complexity of tissues ranges from engineered tissue constructs including a single cell type (i.e. Epiderm™, MatTech, Ashland, Mass., USA) to human skin grafts, to “micro-organs” (i.e. pancreatic islets), to whole human organs (i.e. kidneys, livers, etc.). In the prior art, preservation of these cell systems, as with single cells and monolayers, is typically performed utilizing liquid preservation media. Due to the complex nature of tissues, the shortcomings of liquid-based preservation technologies include those associated with cell and monolayer preservation, along with the inability to maintain the more intricate cell to cell interactions that are stressed during periods of preservation.
  • The gelled preservation media of the present invention enable preservation of tissue in a semi-solid preservation matrix, keeping the tissue structurally intact while affording the same protective benefits to the tissue as is conferred to individual cells and cell monolayers. Specifically, the reduction in the external mechanical shipping forces experienced by the tissues shipped in the inventive gel medium markedly improves tissue viability following preservation. For example, pancreatic islet cells preserved in a medium comprising gelatin and Hypothermosol® were afforded significant improvement of islet integrity and functionality following preservation (see FIG. 9).
  • Types of gellation material suitable for use in a gelled cell preservation medium include, but are not limited to, gelatin, carrageenan, agarose, collagen, laminin, fibronectin, plant-based gelling agents, and combinations thereof. Plant-based gelling agents include gels produced from the roots, stems, tubers, fruit and seeds of plants.
  • The invention is further described in the following non-limiting examples.
  • EXAMPLES Example 1
  • Gel-Based Medium Preparation
  • Calculations are first performed to determine the necessary volume of gel-based medium for a given storage or transport need. The desired gel concentration must be established. Typically, a standard concentration of 2% is used, although this concentration may vary depending upon the characteristics of the biologic being preserved. A stock solution of gel-based medium was then prepared (standard=14%) in a sterile environment. The volume of stock solution needed was determined and the mass of the appropriate amount of gelatin powder (Sigma Chemicals, St. Louis, Mo., USA) was ascertained. The appropriate volume of organ preservation solution (Hypothermosol®, or “HTS”) was measured and combined via agitation with the gelatin powder. In this instance, the stock solution was mixed by swirling the container having the HTS and gelatin, although any suitable means of agitation may be employed. The stock solution of gel-based medium was then warmed in a 40° C. water bath for 15-20 min with repeated swirling solution (once per minute) to dissolve the gelatin. A 2% solution of gel-based medium was then prepared from this stock solution. The desired volume depended upon the quantity of cells to be preserved. For example, for 100 ml of the 2% solution of gel-based medium, 14 ml of 14% stock solution of gel-based medium was combined with 86 ml of an HTS-Free Radical Scavenger (FRS) solution. The FRS solutions discussed herein and shown in the figures include the anti-oxidant Trolox (vitamin E analogue) which is an anti-apoptotic agent. Aliquots of this 2% solution of gel-based medium were dispensed into 15 ml centrifuge tubes at 10 ml per tube, which were stored at 4° C. until used.
  • Example 2
  • Gel-Based Medium Storage Protocol
  • Aliquots of previously prepared 2% gel-based medium were removed from 4° C. storage and placed into a 37° C. water bath for 15 minutes to melt the gelatin. While the gel-based medium was warming, samples destined for preservation were prepared in a sterile environment. The desired number of cells to be preserved was transferred into a clean centrifuge tube and was gently centrifuged to pellet cells. Typically, centrifugation at 500×g for 6 min is sufficient to generate a cell pellet from which a supernatant can be decanted. Pelleted cells were then suspended in 0.5-1.0 ml of HTS-FRS solution without gelatin, which is an appropriate volume for preservation in 2% gel-based HTS-FRS medium. The warmed 2% gel-based medium, now in solution form, was removed from the water bath and the suspended cells were pipetted into the warmed medium in a sterile cell culture environment. After tightly securing lids onto the sample storage tubes, the tubes were immediately placed into an ice-water bath for 5 minutes to allow for rapid solidification of the gel-based medium solution. A 2% gel-based medium solution will solidify around room temperature in approximately 30 min, and chilling was used to accelerate the solidification. Chilled sample tubes were then transferred to a desired storage temperature.
  • Samples prepared in this manner may be stored for any desired time period at appropriate temperatures. For example, cells may be stored for less than 24 hours or for as long as about three days. In one embodiment, storage temperatures preferably range from about −196° C. to about +30° C.
  • Temperatures are considered “normothermic” in a range between 31° C.-37° C. Temperatures are considered “hypothermic” in a range between 0° C. and 30° C. Cryopreservation generally occurs at temperature below 0° C., and may be achieved using a combination of a gel-based media with cryopreservants.
  • Sample tubes are removed from storage when the cells contained therein are to be used. Accordingly, the sample tubes in this instance were placed in a 37° C. water bath for 6-8 minutes to melt the gelled medium. However, the gelling agent can be formulated into a preservation medium such that melting can occur at any desired temperature above or below 37° C. While samples were thawing, tubes were inverted every 30 seconds to maintain uniform temperature throughout the samples. Once the gelled medium containing sample cells melted, samples were immediately removed from the water bath. Cell samples were then gently centrifuged to form pellets, typically at 37° C. at 500×g for 6 min. Cell sensitivity to heat stress will delimit tolerable temperature ranges for gel melting for a given biologic. Centrifugation can be performed at room temperature, but the gel-based medium solution may partially resolidify at this temperature. Resolidification causes uncontrolled gel concentration within the cell pellet during centrifugation. For this reason centrifugation at temperatures between 30° C.-37° C. is preferred.
  • After centrifugation, the gel-based medium supernatant was decanted from the cell pellet, which was then suspended in 12 ml of an appropriate cell culture medium at 37° C. to wash residual gel solution from the cells. The samples were then gently centrifuged to pellet cells at 37° C., typically at 500×g for 6 min. The supernatant was decanted from the pelleted cells prior to resuspension to a desired cell density in an appropriate volume of cell culture media at 37° C. Suspended cells were then transferred to cell culturing vessels at the density desired for growth.
  • FIG. 1 shows post-storage recovery of MDCK (Madin Darby Canine Kidney) cells stored as monolayers in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 1 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • The data in FIG. 1 shows the relative survival of MDCK cells following cold storage in either liquid (media (RPMI culture media), HypoThermosol (HTS), HypoThermosol-FRS (HTS-FRS) or gel-state preservation (HTS-gel; HTS-FRS gel) compared to pre-preservation cultured cells (control). System viability was determined based on the metabolic function of each sample. The data show that MDCK survival following 24 hours of gel-state preservation is equivalent to that of liquid storage demonstrating that gel-state storage is a viable and effective preservation modality. The reduced viability at 1 hour post-storage is due to the depression of metabolism in the system following storage for which a recovery interval is necessary to resume proper function. This recovery period is what is responsible for the differential between the 1 hour and 24 hour data points. Of note, the lower the redound differential (the least change from 1 hour to day 1), the better preserved (less stressed) the cell system.
  • FIG. 2 shows post-storage recovery of MDCK cells stored as monolayers in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 2 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions.
  • The data in FIG. 2 shows the relative survival of MDCK cells following cold storage in either liquid or gel-state preservation compared to pre-preservation cultured cells (control). System viability was determined based on metabolic function of each sample. Overall, the data show that MDCK survival following 3 days of gel-state preservation is equivalent to that of liquid storage demonstrating that gel-state storage is a viable and effective preservation modality. The reduction of viability at 1 hour post-storage is due to the depression of metabolism in the system following storage for which a recovery interval is necessary to resume proper function. This recovery period is what is responsible for the differential between the 1 hour and 24 hour data points. Of note, the lower the redound differential (least change from 1 hour to day 1) the better preserved (less stressed) the cell system.
  • FIG. 3 shows post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 24 hours in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 3 shows post-storage recovery at 1 hour, 1 day and 2 days after the cells have been removed from the 4° C. storage conditions. The use of media only (media, hour data) and HTS (hour data) resulted in a significant loss in cell viability. In contrast, the use of HTSgel (hour data) resulted in a maintenance of cell survival post-storage similar to controls.
  • The data in FIG. 3 demonstrates that gel-state preservation is more effective in that cell systems can be more optimally preserved under similar conditions (see 1 hour data HTS vs. HTS-gel). Interestingly, this increased protection is cell type and formulary specific as a result of the variance in cell death responses (apoptotic and necrotic) activated during and following gel state-preservation (compare 1 hour HTS GEL vs. HTS FRS Gel).
  • FIG. 4 shows post-storage recovery of MDCK cells stored in suspension in culture plates at 4° C. for 3 days in liquid maintenance and preservation solutions (controls) (media, HTS, & HTS FRS) and in liquid maintenance and preservation solutions as gel formulations (HTS Gel & HTS FRS Gel). More specifically, FIG. 4 shows post-storage recovery at 1 hour and 1 day after the cells have been removed from the 4° C. storage conditions.
  • The data in FIG. 4 shows the relative survival of MDCK cells following cold storage in either liquid (media (RPMI culture media), HypoThermosol (HTS), HypoThermosol-FRS (HTS-FRS), or gel-state preservation (HTS-gel; HTS-FRS gel) compared to pre-preservation cultured cells (control). System viability was determined based on the metabolic function of each sample. The data from this series of studies demonstrates the advantage of gel state preservation in reducing the time dependent variability in samples during the recovery period. While overall sample viability is important, consistency during the recovery period following extended storage (such as 3 days) is also critical in cell suspension samples.
  • FIG. 5 graphically depicts post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 1 day in maintenance and preservation solutions as gel formulations. More specifically, FIG. 5 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 22° C. storage conditions.
  • The data in FIG. 5 demonstrates the relative survival of islets following gel-state preservation at room temperature—a thermal range not possible for storage under other preservation regimes. Islets were stored in three gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day and 3 day data). The dip in metabolic activity seen at 2 days post storage in all the stored samples was due to the metabolic cycling within the islet samples as the various cell populations within the islets began to re-synchronize following the preservation process. Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. For example, the xHTS-gel solution and the xHTS-FRS-gel solution shown in this figure differ in that the xHTS-FRS-gel solution includes the antioxidant Trolox; the two solutions are otherwise identical. The HTS-gel solution shown in this figure is composed of approximately two times the concentrations of each of its components than the xHTS-gel solution. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 6 shows post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 22° C. for 3 days in maintenance and preservation solutions as gel formulations. More specifically, FIG. 6 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 22° C. storage conditions.
  • The data in FIG. 6 demonstrates the relative survival of islets following gel-state preservation at room temperature for 3 days—a thermal range not possible for storage under other preservation regimes. Islets were stored in three gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day and 3 day data). In contrast to FIG. 5, no dip in metabolic activity is seen at 2 days post storage. This is due to the synchronization event of the islets occurring during the extended cold storage interval. Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 7 shows post-storage recovery of human pancreatic Islets of Langerhans micro-organs stored in suspension in culture plates at 8° C. for 1 day in maintenance and preservation solutions as gel formulations. More specifically, FIG. 7 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 8° C. storage conditions.
  • The data in FIG. 7 demonstrates the relative survival of islets following gel-state preservation at hypothermic temperatures for 1 day. Islets were stored in two low temperature gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day, 2 day, 3 day data). Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions.
  • FIG. 8 shows post-storage recovery of human pancreatic Islets of Langerhans stored in suspension in culture plates at 8° C. for 3 days in maintenance and preservation solutions as gel formulations. More specifically, FIG. 8 shows post-storage recovery at 1 hour, 1 day, 2 days and 3 days after the cells have been removed from the 8° C. storage conditions.
  • The data in FIG. 8 demonstrates the relative survival of islets following gel-state preservation at hypothermic temperatures for 3 days. Islets were stored in two low temperature gel-based preservation media and compared to non-preserved cultured controls. The data show that following the initial recovery period of the islet systems, the gel state preservation regime was highly effective in maintaining islet viability (1 day, 2 day and 3 day data). Each of the gel-preservation media differed in their composition on the level of ionic, antioxidant, energy substrate, anti-apoptotic agent, impermanent, etc agent concentrations. As such, the data also demonstrate the effectiveness of gel-state preservation over a wide array of base preservation media compositions. Further, these data demonstrate that by storing islets in the cold in gel-state preservation one can maintain islet survival both in the cold and in culture for extended periods (3 days in the cold) and have minimal effect on long term survival (3 days culture) where as simple culture of the islets for 3 days results in a substantial decline in sample viability (decrease in viability in controls over three days).
  • Referring now to FIG. 9, a direct comparison of a media based approach, a HTS based approach and the approach of the present invention utilizing a Human Islet model for storage at 22° C. reveals that both the media and HTS based approaches fail to protect the islets to any degree. In contrast, the gel-based media facilitates substantial protection of the islets during storage, thereby resulting in increased viability post-storage.
  • The data in FIG. 9 demonstrates the comparison between islet survival following storage at 22° C. for 3 days in liquid or gel-state preservation. The data are compared to typical islet survival following liquid storage in culture media (RPMI) for 1 day at 22° C. (industry standard shipment regime). The data show that gel-state preservation and the control of molecular based cell death (using anti-apopotic agents, such as the ant-oxidant Trolox found in the Gelstor X-FRS) results in a significant improvement in cell survival compared to both short-term (Media 1 day) and long-term (media 3 day, HTS, and HTS-FRS) liquid storage. Specifically, in the case of islets, this improvement in metabolic activity (islet function) was seen to be an 80% overall improvement at day 1 following 3 days of gel storage compared to 1 day liquid storage. This represents a significant advancement in preservation efficacy for islets going from one day to 3 days while providing for greater cell viability and function.
  • Gel-Based Preservation Solutions Including One or More Apoptotic Agents
  • The present invention also includes a method of preserving a eukaryotic cell, tissue or organ by contacting the cell, tissue or organ with a hypothermic storage solution containing an agent that inhibits apoptotic cell death and a gelling agent, and storing the cell, tissue or organ and the hypothermic storage solution surrounding or perfusing the cell, tissue or organ. The gel-state preservation is accomplished through use of a hypothermic storage solution that has a gelling agent. Reduction of the temperature of a sample in such a solution to below the gelling temperature results in the gel-state encapsulation of the cell, tissue or organ in that solution. The inclusion of one or more anti-apoptotic agents aids in preventing the apoptotic cell death that normally occurs subsequent to this type of preservation.
  • Gel State Preservation:
  • Gel-state preservation according to the invention involves the use of a hypothermic storage solution that forms a semi-solid matrix (gel), rather than a liquid or crystalline solid when exposed to low temperatures, specifically, temperatures below the gelling point of the solution. Gel-state preservation as the term is used herein involves the non-crystalline liquid to semi-solid phase transition of the surrounding or perfused hypothermic storage solution thereby encapsulating the cell, tissue, or organ. This gel-state thus does not involve the formation of macromolecular crystals (such as ice crystals) within or outside of cells, tissues or organs placed into or perfused with such a solution. This is in contrast to cryopreservation, in which ice crystals are permitted to form outside of such cells, or hypothermic storage where cells are bathed in a liquid media for the preservation interval.
  • In one embodiment, gel-state preservation involves the steps of contacting the cells, tissue or organ with the hypothermic storage solution that contains one or more agents that inhibit apoptosis, and then reducing the temperature below the gelling point (solidification point) of the solution, such that a gel-like semi-solid is formed. The rate of cooling depends upon the solution, but is generally in the range of 0.5° C. to 50° C. per minute, most often approximately 1° C. to 20° C. per minute, e.g., approximately 1° C. per minute, approximately 5° C. per minute, approximately 10° C. per minute, approximately 15° C. per minute or approximately 20° C. per minute. The skilled artisan can identify an optimal rate of cooling for a given combination of the gel-state solution and tissue.
  • Gel-state formation may be monitored in several ways. One of the simplest ways is to observe the solution (solid vs. liquid above 0° C.) or by observing or measuring the transmission of light through the gel.
  • The gel-state sample is then stored below the gelling point. The gelling point generally ranges from +25° C. to −196° C. (liquid nitrogen temperature). Further reduction in temperature below the gelling point is often desirable. Such preservation maintains the semi-solid state of the gel without permitting melting or crystallization within and outside of cells.
  • The step of contacting cells, tissues or organs with a hypothermic storage solution differs depending upon whether the sample is cells, a tissue or an organ. For example, cells or relatively thin (e.g., up to about 2-5 mm) or porous tissues (e.g., skin or artificial skin) can simply be immersed in the hypothermic storage solution prior to chilling to cause gelling. This immersion is preferably performed at or near 22° C. or 4° C. The length of time necessary for immersion will clearly vary with the thickness of the tissue, with longer times necessary as the tissue becomes thicker. One of skill in the art can readily evaluate and adjust the time necessary for the immersion of a given sample or type of sample by evaluating the survival, recovery and degree of apoptosis occurring in samples immersed for varying lengths of time before the initiation of gelling.
  • For organs, e.g., kidney, liver, heart, lung, etc., it may be necessary to perfuse the organ with the hypothermic storage solution. Methods for organ perfusion vary for different organs but are well known in the art. For perfusion of organs, an important parameter is the viscosity of the solution. More porous organ tissues (e.g., liver) may be perfused with relatively higher viscosity solutions than can less porous tissues (e.g., heart). Because of differences in the ability to perfuse different tissues, it may be necessary in some cases to perfuse with an increasing gradient, or with stepwise increases, of the concentration of hypothermic storage solution until a concentration is achieved such that the tissue will gel throughout upon subsequent reduction of temperature.
  • Apoptosis and Anti-Apoptotic Agents:
  • The precise cellular mechanisms regulating apoptosis are not completely known. However, various aspects of the apoptotic pathway have been elucidated. For example, alteration of the ionic environment may be necessary to activate or inhibit the endonucleases relevant to the process of apoptotic nuclear degradation (e.g., physiologic concentrations of Zn++ are known to inhibit DNA fragmentation and apoptosis). Treatment of certain cells with inhibitors of macromolecular synthesis, such as Actinomycin D to block RNA synthesis or cyclohexamide to block protein synthesis, induces apoptosis. Completion of the apoptotic process appears to depend upon the regulated expression of various gene products associated with the promotion or suppression of gene activated cell death, particularly gene products involved with cell cycle regulation. For example, overexpression of the cell-death inhibiting agents Bcl-2 and Bcl-xL prevents the release of cytochrome C. Cytochrome C is thought to activate the caspases, a group of proteases known for cleaving substrates responsible for the changes associated with apoptosis. Enhanced levels of Bax, a pro-apoptotic member of the Bcl-2 family, promote cytochrome C release and subsequent apoptosis of cells. Specific regulation of the early response genes c-myc, c-jun and c-fos may promote either cell growth or cell death, depending upon the circumstances surrounding their expression. Another trigger for apoptosis involves oxidative stress. In this regard, antioxidants and free-radical scavengers have been demonstrated to inhibit the initiation of apoptosis. Thus, programmed cell death involves an intricate cascade of cellular events.
  • The inventors of the present invention have determined that hypothermic storage is among the numerous triggers for apoptosis. In particular, the gel-state preservation of cells tends to cause a portion of the cells to undergo apoptosis and/or necrosis when they are removed from gel-state storage. While it is not known exactly what molecular mechanism is the triggering event for gel preservation-induced apoptosis, the inclusion, in the hypothermic storage solution used for gel-state preservation, of one or more agents that alone or collectively inhibit apoptosis can increase the survival and recovery of the cells.
  • There are a number of agents known to inhibit apoptosis. Agents useful according to the invention can function at any stage of the apoptotic pathway, e.g., by modulating the function or expression of one or more nuclear or cytoplasmic polypeptide mediators of the pathway (e.g., gene products in the regulatory cascade such as Bcl-xL, Bcl-2 or Bax, cytochrome C, caspase enzymes, etc.) or by avoiding or countering the effects of oxidative stress known to trigger apoptosis (e.g., through use of antioxidants, free radical scavengers or agents that modulate the function or expression of nitrous oxide synthase).
  • One useful way to categorize inhibitors of apoptosis is to consider those that interact with a cellular polypeptide factor that participates in the generation, propagation or execution of an apoptotic signal to be one category, and those that inhibit by other means as a second category. In the first category, the interaction of the agent with the polypeptide factor is a physical, i.e., binding, interaction. For example, an agent that physically interacts with and inhibits the function of a caspase enzyme would fall into this category. In the second category the agent need not necessarily interact with or bind a polypeptide, but rather acts by, for example scavenging free radicals or maintaining the redox status quo. Examples of agents in the second category include, for example, Vitamin E and other agents listed in Table II.
  • According to an embodiment of the present invention, a composition that inhibits apoptosis includes at least one agent that reduces the extent of apoptotic cell death, but may include, for example, two or more such agents, where each such agent preferably, but not necessarily, interacts with a different part of an apoptotic pathway. In another embodiment, the composition includes two or more agents that together have an inhibitory effect on apoptosis that neither has on its own, or that together have a synergistic effect.
  • An anti-apoptotic agent useful according to the invention will inhibit apoptosis, as measured by a TUNEL assay, by at least 10% relative to a sample subjected to the same apoptotic induction conditions without the addition of the anti-apoptotic agent.
  • Some cellular polypeptide targets involved in the promotion of apoptosis are listed in Table IA. In one embodiment of the present invention, an anti-apoptotic agent useful according to the invention can act on one or more of these cellular polypeptide targets. Table IB also lists exemplary targets involved in the prevention of apoptosis. In another embodiment, an anti-apoptotic agent can act to increase or stabilize the activity of one or more of these cellular polypeptide targets.
  • As noted, an important class of anti-apoptotic agents is the caspase inhibitors, a number of which are known and commercially available. One class of caspase inhibitors comprises fluoromethylketone (FMK) derivatives of peptides that mimic the recognition and cleavage sites for the target caspase enzymes. Among these are, for example: caspase-1 inhibitor YVAD-FMK (benzyloxycarbonyl tyrosylvalylalanyl aspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 1; caspase-3 inhibitor DEVD-FMK (benzyloxyycarbonyl aspartyl glutamylvalylaspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 3 but also inhibits caspase 7, caspase 10, and caspase 6 in decreasing order of binding affinity; caspase-6 inhibitor VEID-FMK (benzyloxycarbonyl valylglutamyl isoleucylaspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 6 and also inhibits caspases 3, 7, and 8 in decreasing order of binding affinity; caspase-8 inhibitor LETD-FMK (benzyloxycarbonyl leucylglutamylthreonylaspartic acid fluoromethyl ketone), which irreversibly binds activated caspase 8, and also binds caspase 1 and caspase 10 with lower affinity; caspase-9 inhibitor LEHD-FMK (benzyloxycarbonyl leucylglutamylhistidylaspartic acid fluoromethyl ketone), which irreversibly binds caspase 9 and also binds caspases 4, 5 and 6. Additional FMK-peptide derivatives useful for the inhibition of caspases include, but are not limited to caspase-2 inhibitor VDVAD-FMK, caspase-4 inhibitor Z-LEVD-FMK, caspase-5 inhibitor WEHD-FMK, caspase-10 inhibitor AEVD-FMK and caspase-13 inhibitor LEED-FMK. These caspase inhibitors, and cocktail mixtures of them are commercially available from, for example, Gentaur Molecular Products (Brussels, Belgium).
  • Additional anti-apoptotic agents include the calpain inhibitors (e.g., Calpain Inhibitor Z-LLT-FMK) and the cathepsin B&L inhibitor Z-Phe-Phe-FMK. These inhibitors are also available from Gentaur Molecular Products. Others include Calpain Inhibitor I (N-Acetyl-Leu-Leu-norleucinal, available from Roche Diagnostics), Calpain Inhibitor II (N-Acetyl-Leu-Leu-methioninal, available from Roche Diagnostics), and CTX295 (a dipeptide alpha-ketoamide compound which inhibits calpain (DiBiasi et al., 2001, J. Virol. 351-361)). Additional anti-apoptotic agents useful according to the invention are listed in Table II.
  • The amount of an anti-apoptotic agent or agents useful in a gel-based storage solution according to the invention will vary depending on the nature of the inhibitor and its target, if the target is known (there is no requirement that the specific target of the apoptotic inhibitor useful according to the invention be known). Generally, the amount of anti-apoptotic agent useful will be that amount or concentration determined by the user to provide at least 10% (and preferably greater) inhibition in apoptosis following removal from the gelled state relative to a sample that was in the gelled state in the absence of the inhibitor or inhibitors. The concentration of anti-apoptotic agent useful according to the invention will further not be associated with significant (i.e., greater than 2% cell death over the course of treatment or contact) toxic effects. The level at which an agent becomes toxic will vary with the agent and will generally be known or readily determined by one of skill in the art.
    TABLE I
    Pathway Location
    (Initiation,
    Execution,
    Target Definition Termination)
    A. Cellular Targets Involved in the Promotion of Apoptosis
    which can be Inhibited to Improve Gel-State Preservation
    Caspases (Cystine Proteases) Initiation,
    Execution,
    Termination
    ROCK
    CAD (Caspase Activated Termination
    DNAse)
    ASK1 Initiation,
    Execution
    JNK (Jun Kinase Family) Initiation
    Fas Initiation
    FADD (Fas Activated Death Initiation
    Domain)
    TNF (Tumor Necrosis Initiation
    Factor)
    TRADD (TNF Receptor Activated Initiation
    Death Domain)
    RIP (receptor Interacting Initiation
    Protein)
    DAXX Initiation
    Granzyme B Initiation
    Bad (Mitochondrial Pro- Initiation,
    apoptotic protein) Execution
    Bax (Mitochondrial Pro- Initiation,
    apoptotic protein) Execution
    Bid Initiation,
    Execution
    Cytochrome C Initiation,
    Execution
    AIF (Apoptosis Initiation Initiation,
    Factor) Execution
    MAPK (Mitogen Activated Initiation
    Protein Kinase Family)
    Calpain (Serine Proteases) Initiation,
    Execution,
    Termination
    Caspathin Initiation,
    Execution,
    Termination
    Nitric Oxide Initiation
    PARP (Poly-ADP Ribose Termination
    Polymerase)
    DFF (DNA Fragmentation Termination
    Factor)
    B. Cellular Targets Involved in the Prevention of Apoptosis
    which can be Activated to Improve Preservation Efficacy
    Bcl-2 (Mitochondrial Anti- Initiation,
    apoptotic protein) Execution
    Bcl-x (Mitochondrial Anti- Initiation,
    apoptotic protein) Execution
    IAP (Inhibitor of Apoptosis Initiation,
    Protein) Execution
    RAS Receptor mediated pro- Initiation
    survival signal
    AKT Anti-apoptosis signal Initiation,
    Execution
    TRAF2 (TNF Receptor Associated Initiation
    Factor 2)
  • TABLE II
    Free Radical Scavengers and Other Anti-apoptotic Agents
    Flavonoids
    Vitamin E
    Vitamin C
    Vitamin D
    Beta Carotene (Vitamin A)
    Pycnogenol
    Super Oxidedismutase
    N-Acetyl Cysteine
    Selenium
    Catechins
    Alpha Lipoic Acid
    Melatonin
    Glutathione
    Zinc Chelators
    Calcium Chelators
    L-Arginine
  • Measurement of Apoptosis:
  • There are a number of ways for one to measure the extent of apoptosis occurring following the gel state preservation of a sample containing cells. As noted, one of the hallmarks of apoptosis is a regular DNA fragmentation pattern leading to a “ladder” when genomic DNA is subjected to gel electrophoresis. This assay is well known in the art, and can be performed as described in U.S. Pat. No. 6,045,990.
  • Another assay useful for more quantitative measurement of apoptosis is the TUNEL assay (Terminal deoxynucleotidyl transferase mediated dUTP nick end labeling). This assay measures the enzymatic incorporation of labeled dUTP at the nick or breaks in DNA that accompany apoptosis. The assay is well known to those of skill in the art, and kits for performing the assay are available, for example, from Intergen (ApopTag™ kit, Intergen Company, Purchase, N.Y.).
  • Alternatively, one may directly assay caspase activity. Kits for this are also commercially available, e.g., from Promega (Madison, Wis.).
  • Other assays for apoptosis are also known to those of skill in the art. For example, Annexin V binding to cells can be measured. One of the early events of apoptosis is the loss of membrane asymmetry of phospholipids. Phosphotidylserine, normally located in the inner leaflet of the membrane, redistributes and appears in the outer leaflet at the early stage of apoptosis. Annexin V binds specifically to phosphotidylserine on apoptotic cell surfaces in the presence of calcium, and can be used as a marker for apoptosis. Binding can be measured by monitoring a fluorescent tag on the Annexin V, e.g., fluorescein.
  • Removal from the Gel State
  • The process for the removal of cells, tissues or organs from the gel-state state will vary depending upon the cell, tissue or organ preserved. The general process involves warming the sample above the gelling point, followed by rinsing or dilution of the sample to remove the hypothermic storage solution, followed by re-establishment in culture or by implantation into a recipient. Rinsing can be performed by simple sample dilution (e.g. for isolated cells and cell suspensions), immersion (e.g., for cells or relatively thin tissues) or by perfusion (e.g., for organs). While minimizing time out of standard culture conditions or a recipient individual is always desirable, it may be advantageous to remove gel solution by stepwise immersion or perfusion with gradually reduced concentrations of solution agents. In such circumstances, it may be advantageous to include an anti-apoptotic composition in the rinse solution or solutions. In fact, an anti-apoptotic composition can advantageously be added to any such rinse solution.
  • Measurement of Gel-State Storage Efficiency:
  • The term “gel-state storage” encompasses storage for a matter of hours or for a matter of weeks, months or years. The level of cell survival acceptable for such storage will depend upon the cell, tissue or organ type stored and the reason for its storage (e.g., transplantation versus research uses), but will advantageously be higher than is practicable using prior art technologies. When a cell, tissue or organ is used for human transplantation, it is advantageous to maximize the survival and ensuing function of the cell, tissue or organ. Generally, however, gel-state storage is considered successful if it is accompanied by death of about 20% or fewer of the cells, preferably 10% or less, and most preferably less than 5%, up to and including full viability and function.
  • The storage efficacy of numerous cell preservation solutions can be assessed using a number of assays. Such assays include enzyme synthesis (Pahernik et al., 1996, Cryobiology 33: 552), potassium content (Fisher et al., 1996, Cryobiology 33: 163), trypan blue exclusion (Rodriguez et al., 1995, Cell Transplant. 4: 245), neuronal outgrowth and myelination (Levi et al., 1994, Glia 10: 121), contraction (Lopukhin et al., 1996, Cryobiology 33: 178), ATP content (Zhang et al., 1996, J. Surg. Res. 63: 314), and ultrastructure (Carbognani, et al., 1995, J. Cardiovasc. Surg. 36: 93). These assays can be considered either viability or functional assays. Thus, the maltose tolerance test used by Katz et al. (Katz et al., 1995, Transplantation 59: 694) on the small intestine could be considered a functional assay; whereas the trypan blue test used by Rodriguez et al. (Rodriguez et al., 1995, supra) is strictly a viability assay. The references cited in this paragraph are all incorporated herein by references.
  • A preferred assay for viability uses the alamarBlue® non-toxic dye. The assay using alamarBlue® dye measures viability by the metabolic conversion of the agent to a fluorescent form detectable with standard fluorescence detection equipment. The assay does not indicate whether or not test cells are functioning in a tissue specific manner. However, one of the key attributes of the alamarBlue® dye assay that is not shared by most of the other viability or function assays described previously is the ability of alamarBlue® dye to be used repetitively, day after day, as a non-toxic indicator. This has been shown to be critically important to some cold-stored tissues such as human skin cells.
  • An example of the conditions for alamarBlue® dye viability testing following gel-state storage is as follows. Cells (e.g., cultured cells or cells isolated from a patient) are washed with isotonic buffer solution (e.g., phosphate buffered saline, Hanks' Balanced Salt Solution (HBSS), etc.) and the medium is replaced with a gel-state storage solution, containing at least one apoptosis inhibiting agent. The cell sample is cooled and maintained for a given time period at low temperature, e.g., 4° C., after which the sample is warmed such that the solution returns to the liquid state. The gel-state storage solution is removed, cells are washed in isotonic buffer solution or cell culture medium and plated for growth in appropriate cell culture medium. After a given period of time (generally about 24 hours, but advantageously later, e.g., 48 hours or more) medium is removed from the recovered cultures, and a 1:20 dilution of alamarBlue® dye (Accumed International, Westlake, Ohio) in HBSS without phenol red is placed on the cells. Fluorescence of the converted dye is measured using, for example, a CytoFluor 2350 (Millipore Corporation, Bedford, Mass.) or a CytoFluor II (PerSeptive Biosystems, Inc., Cambridge, Mass.) apparatus with a 530 nm excitation/590 nm emission filter set. Following the assay, cells can be provided with fresh medium and re-cultured with repeat assays at later times as desired.
  • Another way to monitor the viability of recovered cells, tissues or organs uses the dye Calcein-AM (Molecular Probes, Eugene, Ore.), which monitors membrane integrity. Calcein-AM is suspended at 1 mg/ml in DMSO and then mixed with HBSS at 1:100. This solution is then contacted with recovered cells for one hour, followed by washing with HBSS. Fluorescence of the retained dye, indicative of dead cells, is then measured using a 485 nm excitation/530 nm emission filter set.
  • The present invention preserves various organs, tissues and cells using semi-solid gel-state storage. Organs, including but not limited to lung, liver, heart, kidney, gut, eye and skin can be preserved according to the invention prior to transplantation in a recipient patient. Tissues such as bone marrow and cells such as erythrocytes and leukocytes can be preserved for long term storage according to the invention. For example, tissues for forensic and pathology records may be preserved without significant loss of viability. Cell lines for therapeutic and research interests can be preserved for long periods by applying the invention. It is contemplated that variations of the invention can be applied for long term preservation of entire multicellular organisms.
  • Accordingly, it is to be understood that the embodiments of the invention herein described are merely illustrative of the application of the principles of the invention. Reference herein to details of the illustrated embodiments is not intended to limit the scope of the claims, which themselves recite those features regarded as essential to the invention.

Claims (45)

1. A method of preserving a eukaryotic cell, tissue or organ comprising:
a) contacting the cell, tissue or organ with a storage solution, wherein the solution comprises:
i) a composition that inhibits apoptosis; and
ii) a concentration of a gelling agent that is sufficient for gel solidification of the solution; and
b) storing the cell, tissue or organ in the solution.
2. The method of claim 1 wherein the storage solution has a gelling temperature below 37° C.
3. The method of claim 1 wherein the gelling agent is selected from the group consisting of:
a) gelatin;
b) carrageenan;
c) agarose;
d) collagen;
e) laminin;
f) fibronectin;
g) a plant based gelling agent; and
h) any combination of a) through g).
4. The method of claim 1 wherein the cell, tissue or organ is stored in the storage solution at a temperature between 30° C. and −196° C.
5. The method of claim 1, wherein the cell, tissue or organ is stored in the storage solution at a temperature between 0° C. and −196° C.
6. The method of claim 1, wherein the cell, tissue or organ is stored below a gelling temperature of the storage solution.
7. The method of claim 1 wherein the composition that inhibits apoptosis comprises an agent that interacts with a polypeptide that participates in an apoptotic pathway.
8. The method of claim 7 wherein the agent inhibits the activity of the polypeptide.
9. The method of claim 7 wherein the agent maintains or potentiates the activity of the polypeptide.
10. The method of claim 7 wherein the agent is selected from the group consisting of a caspase inhibitor, a calpain inhibitor, and an inhibitor of nitrous oxide synthase.
11. The method of claim 1 wherein the composition that inhibits apoptosis comprises an antioxidant.
12. The method of claim 1 wherein the composition that inhibits apoptosis comprises an agent selected from the group consisting of a free radical scavenger, a zinc chelator, and a calcium chelator.
13. The method of claim 1, wherein the storage solution further comprises at least one agent that protects the cells, tissue, or organ from ice-related damage.
14. The method of claim 13, wherein the agent that protects the cells, tissue, or organ from ice-related damage is selected from the group consisting of dimethyl sulfoxide, glycerol, and propanediol.
15. A preservation solution comprising:
a) a composition that inhibits apoptosis, and
b) a gelling composition that comprises a concentration of one or more agents that is sufficient for gel solidification of the solution when the temperature of the solution is reduced below the gelling point of the solution.
16. The solution of claim 15 wherein the gelling point of the solution is higher than the homogeneous nucleation temperature of the solution.
17. The solution of claim 15 wherein the composition that inhibits apoptosis comprises an agent that interacts with a polypeptide that participates in an apoptotic pathway.
18. The solution of claim 17 wherein the agent inhibits the activity of the polypeptide.
19. The solution of claim 17 wherein the agent maintains or potentiates the activity of the polypeptide.
20. The solution of claim 17 wherein the agent is selected from the group consisting of a caspase inhibitor, a calpain inhibitor, and an inhibitor of nitrous oxide synthase.
21. The solution of claim 20, wherein the agent is a caspase inhibitor and is selected from the group consisting of peptide fluoromethyl ketone, CHO, a peptide chloromethyl ketone, DCB, AOM and FAOM.
22. The solution of claim 20 wherein the agent is a calpain inhibitor and is selected from the group consisting of leupeptin, calpain inhibitors I, II, III, IV and V, calpeptin, loxastatin, a peptide chloromethyl ketone and a peptide fluoromethyl ketone.
23. The solution of claim 16 wherein the composition that inhibits apoptosis comprises an antioxidant.
24. The solution of claim 23 wherein the antioxidant is selected from the group consisting of glutathione, N-acetyl cysteine, beta carotene, Vitamins E, D, C and A, Nitric Oxide, L-arginine, and super oxide dismutase.
25. The solution of claim 16, wherein the composition that inhibits apoptosis comprises an agent selected from the group consisting of a free radical scavenger, a zinc chelator, and a calcium chelator.
26. The solution of claim 25, wherein the agent is a free radical scavenger selected from the group consisting of Vitamins E, D, C and A, Nitric oxide, L-arginine and super oxide dismutase.
27. The solution of claim 16, further comprising at least one agent that protects the cells, tissue, or organ from ice-related damage.
28. The solution of claim 27, wherein the agent that protects the cells, tissue, or organ from ice-related damage is selected from the group consisting of dimethyl sulfoxide, glycerol, and propanediol.
29. A method of preserving a eukaryotic cell, tissue or organ comprising:
a) contacting the cell, tissue or organ with a storage solution, wherein the solution comprises:
i) a composition that inhibits apoptosis; and
ii) a concentration of a gelling agent that is sufficient for gel solidification of the solution; and
b) storing the cell, tissue or organ in a gel-state wherein the gel-state occurs in the storage solution comprising and comprised by the cell, tissue or organ thereby encapsulating the cell, tissue, or organ.
30. A gel-based medium composition for transport or storage of cell samples, the composition comprising:
(a) a plurality of electrolytes comprising potassium ions at a concentration ranging from about 10-145 mM, sodium ions at a concentration ranging from about 10-120 mM, and calcium ions at a concentration ranging from about 0.01-1.0 mM;
(b) a macromolecular oncotic agent having a size sufficiently large to limit escape from the circulation system and effective to maintain oncotic pressure equivalent to that of blood plasma and selected from the group consisting of human serum albumin, polysaccharide and colloidal starch;
(c) a biological pH buffer effective under physiological and hypothermic conditions;
(d) a nutritive effective amount of at least one simple sugar;
(e) an impermeant and hydroxyl radical scavenging effective amount of mannitol;
(f) an impermeant anion impermeable to cell membranes and effective to counteract cell swelling during cold exposure, the impermeant ion being at least one member selected from the group consisting of lactobionate, gluconate, citrate and glycerophosphate-like compounds;
(g) a substrate effective for the regeneration of ATP, the substrate being at least one member selected from the group consisting of adenosine, fructose, ribose and adenine;
(h) at least one agent which regulates cellular levels of free radicals; and
(i) at least one gelling agent.
31. The gel-based medium composition according to claim 30, further comprising at least one agent which inhibits apoptosis.
32. The gel-based medium composition according to claim 30, wherein the gelling agent is selected from the group consisting of gelatin, carrageenan, agarose, collagen and plant-based gelling agents.
33. The gel-based medium composition according to claim 30, wherein the transport or storage of cell samples occurs at a temperature ranging from about −196° C. to about 37° C.
34. The gel-based medium composition according to claim 30, wherein the cell samples are obtained from a source selected from the group consisting of plants, animals, fungi, and microbes.
35. The gel-based medium composition according to claim 34, wherein the source of the cell samples is a fetal, neonatal, juvenile or adult animal.
36. The gel-based medium composition according to claim 34, wherein the cell samples are obtained from humans.
37. The gel-based medium composition according to claim 30, the plurality of electrolytes further comprising magnesium ions at a concentration ranging from about 0.1-10 mM.
38. The gel-based medium composition according to claim 30, wherein the gel-based composition preserves the cell samples when cooled to a chilled or frozen state.
39. The gel-based medium composition according to claim 30, wherein the cell samples are selected from the group consisting of a plurality of pancreatic islet cells; a plurality of stem cells; and a plurality of liver cells.
40. A method for increasing a storage duration of a cell sample at frozen temperatures, the method comprising:
a) forming a cell pellet;
b) mixing the pellet with a cell preservation medium comprising a cryopreservant, the cell preservation medium not containing a gelling agent;
c) adding a medium comprising a gelling agent to the cell pellet, the medium comprising the gelling agent being in a liquid state when added; and
d) reducing the temperature of the resulting mixture until the mixture gels; and storing the gelled mixture at a temperature below 0° C.;
wherein at least one agent that inhibits apoptosis is part of the medium selected from the group consisting of the cell preservation medium; the medium comprising the gelling agent; and both the cell preservation medium and the medium comprising the gelling agent.
41. The method of claim 40 wherein the gelled mixture is stored at a temperature of below 0° C. to −196° C.
42. The method of claim 40 wherein the cell sample is obtained from a source selected from the group consisting of plants, animals, fungi and microbes.
43. The method of claim 40 wherein the source of the cell sample is a fetal, neonatal, juvenile or adult animal.
44. The method of claim 40 wherein the cell sample is a human cell sample.
45. The method of claim 40 wherein the cell preservation medium comprises one or more electrolytes selected from the group consisting of potassium ions at a concentration ranging from about 10-145 mM, sodium ions ranging from about 10-120 mM, magnesium ions ranging from about 0.1-10 mM, and calcium ions ranging from about 0.01-1.0 mM.
US11/553,245 2000-01-14 2006-10-26 Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State Abandoned US20070048726A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/553,245 US20070048726A1 (en) 2000-01-14 2006-10-26 Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US17600900P 2000-01-14 2000-01-14
US09/757,694 US6632666B2 (en) 2000-01-14 2001-01-11 Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media
US10/166,732 US20020172934A1 (en) 2000-01-14 2002-06-12 Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media
US11/553,245 US20070048726A1 (en) 2000-01-14 2006-10-26 Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US09/757,694 Division US6632666B2 (en) 2000-01-14 2001-01-11 Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media
US10/166,732 Continuation-In-Part US20020172934A1 (en) 2000-01-14 2002-06-12 Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media

Publications (1)

Publication Number Publication Date
US20070048726A1 true US20070048726A1 (en) 2007-03-01

Family

ID=37804671

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/553,245 Abandoned US20070048726A1 (en) 2000-01-14 2006-10-26 Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State

Country Status (1)

Country Link
US (1) US20070048726A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050276728A1 (en) * 2004-04-08 2005-12-15 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20060099567A1 (en) * 2004-04-08 2006-05-11 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20080176209A1 (en) * 2004-04-08 2008-07-24 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20080268514A1 (en) * 2007-04-24 2008-10-30 Rolf Muller Sample storage for life science
US20090239208A1 (en) * 2008-03-21 2009-09-24 Veronique Mayaudon Red Blood Cell Storage Medium For Extended Storage
WO2009121002A1 (en) * 2008-03-27 2009-10-01 Biolife Solutions, Inc. Materials and methods for hypothermic collection of whole blood
WO2010064054A1 (en) * 2008-12-05 2010-06-10 Reneuron Limited Cellular compositions for use in therapy
ES2343721A1 (en) * 2008-12-19 2010-08-06 Histocell, S.L. Cell transport system
US20110117647A1 (en) * 2008-03-21 2011-05-19 Fenwal, Inc. Red Blood Cell Storage Medium For Extended Storage
US9345857B2 (en) 2009-04-29 2016-05-24 Cardio3 Biosciences Injection catheter for delivering a therapeutic agent into a substrate
US9376709B2 (en) 2010-07-26 2016-06-28 Biomatrica, Inc. Compositions for stabilizing DNA and RNA in blood and other biological samples during shipping and storage at ambient temperatures
US9409128B2 (en) 2009-10-23 2016-08-09 Fenwal, Inc. Methods for storing red blood cell products
US9410122B2 (en) 2009-02-06 2016-08-09 Reneuron Limited Treatment of limb ischemia
US9725703B2 (en) 2012-12-20 2017-08-08 Biomatrica, Inc. Formulations and methods for stabilizing PCR reagents
US9845489B2 (en) 2010-07-26 2017-12-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA and proteins in saliva and other biological samples during shipping and storage at ambient temperatures
CN108477143A (en) * 2018-03-27 2018-09-04 福建三造血技术有限公司 A kind of cell transport holding liquid and transportation resources
US10064404B2 (en) 2014-06-10 2018-09-04 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US10568317B2 (en) 2015-12-08 2020-02-25 Biomatrica, Inc. Reduction of erythrocyte sedimentation rate
CN114586768A (en) * 2020-12-03 2022-06-07 江苏齐氏生物科技有限公司 Liver tissue preservation solution and preservation method and application thereof

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559298A (en) * 1982-11-23 1985-12-17 American National Red Cross Cryopreservation of biological materials in a non-frozen or vitreous state
US4774093A (en) * 1985-06-25 1988-09-27 Fmc Corporation Polysaccharide compositions, preparation and uses
US4840891A (en) * 1986-09-03 1989-06-20 Genetic Engineering, Inc. Encapsulation of sperm for artificial insemination
US5217860A (en) * 1991-07-08 1993-06-08 The American National Red Cross Method for preserving organs for transplantation by vitrification
US5328821A (en) * 1991-12-12 1994-07-12 Robyn Fisher Cold and cryo-preservation methods for human tissue slices
US5405742A (en) * 1993-07-16 1995-04-11 Cyromedical Sciences, Inc. Solutions for tissue preservation and bloodless surgery and methods using same
US5498427A (en) * 1990-11-20 1996-03-12 Pasteur Merieux Serums Et Vaccines Solutions for the perfusion, preservation and reperfusion of organs
US5554527A (en) * 1989-11-24 1996-09-10 Behringwerke Aktiengesellschaft Agent for the storage and suspension of cells, especially erythrocytes
US5635344A (en) * 1994-12-07 1997-06-03 Cedra Corp. Shipping medium for organ-derived cells
US5635187A (en) * 1993-11-30 1997-06-03 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
US5723282A (en) * 1991-07-08 1998-03-03 The American National Red Cross Method of preparing organs for vitrification
US6045990A (en) * 1998-07-09 2000-04-04 Baust; John M. Inclusion of apoptotic regulators in solutions for cell storage at low temperature
US6248588B1 (en) * 1996-03-12 2001-06-19 Aventis Pharms S.A. Medium for preserving biological materials
US6303388B1 (en) * 1995-03-30 2001-10-16 Organ Recovery Systems, Inc. Process for preparing novel ice-controlling molecules
US6395467B1 (en) * 1998-09-21 2002-05-28 Gregory M. Fahy Cryoprotectant solution containing dimethyl sulfoxide, an amide and ethylene glycol
US6413713B1 (en) * 1998-10-30 2002-07-02 Hyperbaric Systems Method for preserving blood platelets
US6632666B2 (en) * 2000-01-14 2003-10-14 Biolife Solutions, Inc. Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media
US20040096813A1 (en) * 2002-11-18 2004-05-20 Biolife Solutions, Inc. Methods and compositions for the preservation of cells, tissues or organs in the vitreous state

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559298A (en) * 1982-11-23 1985-12-17 American National Red Cross Cryopreservation of biological materials in a non-frozen or vitreous state
US4774093A (en) * 1985-06-25 1988-09-27 Fmc Corporation Polysaccharide compositions, preparation and uses
US4840891A (en) * 1986-09-03 1989-06-20 Genetic Engineering, Inc. Encapsulation of sperm for artificial insemination
US5554527A (en) * 1989-11-24 1996-09-10 Behringwerke Aktiengesellschaft Agent for the storage and suspension of cells, especially erythrocytes
US5498427A (en) * 1990-11-20 1996-03-12 Pasteur Merieux Serums Et Vaccines Solutions for the perfusion, preservation and reperfusion of organs
US5723282A (en) * 1991-07-08 1998-03-03 The American National Red Cross Method of preparing organs for vitrification
US5217860A (en) * 1991-07-08 1993-06-08 The American National Red Cross Method for preserving organs for transplantation by vitrification
US6187529B1 (en) * 1991-07-08 2001-02-13 The American National Red Cross Method for preparing organs for transplantation after cryopreservation
US5962214A (en) * 1991-07-08 1999-10-05 The United States Of America As Represented By The American National Red Cross Method of preparing tissues and cells for vitrification
US5821045A (en) * 1991-07-08 1998-10-13 The American National Red Cross Methods for removal of cryoprotectant from organs prior to transplantation
US5328821A (en) * 1991-12-12 1994-07-12 Robyn Fisher Cold and cryo-preservation methods for human tissue slices
US5405742A (en) * 1993-07-16 1995-04-11 Cyromedical Sciences, Inc. Solutions for tissue preservation and bloodless surgery and methods using same
US5514536A (en) * 1993-07-16 1996-05-07 Cryomedical Sciences, Inc. Solutions for tissue preservation and bloodless surgery and methods using same
US5635187A (en) * 1993-11-30 1997-06-03 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
US5635344A (en) * 1994-12-07 1997-06-03 Cedra Corp. Shipping medium for organ-derived cells
US6303388B1 (en) * 1995-03-30 2001-10-16 Organ Recovery Systems, Inc. Process for preparing novel ice-controlling molecules
US6248588B1 (en) * 1996-03-12 2001-06-19 Aventis Pharms S.A. Medium for preserving biological materials
US6045990A (en) * 1998-07-09 2000-04-04 Baust; John M. Inclusion of apoptotic regulators in solutions for cell storage at low temperature
US6395467B1 (en) * 1998-09-21 2002-05-28 Gregory M. Fahy Cryoprotectant solution containing dimethyl sulfoxide, an amide and ethylene glycol
US6413713B1 (en) * 1998-10-30 2002-07-02 Hyperbaric Systems Method for preserving blood platelets
US6632666B2 (en) * 2000-01-14 2003-10-14 Biolife Solutions, Inc. Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media
US20040096813A1 (en) * 2002-11-18 2004-05-20 Biolife Solutions, Inc. Methods and compositions for the preservation of cells, tissues or organs in the vitreous state
US6921633B2 (en) * 2002-11-18 2005-07-26 Biolife Solutions Incorporated Methods and compositions for the preservation of cells, tissues or organs in the vitreous state

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090291427A1 (en) * 2004-04-08 2009-11-26 Judy Muller-Cohn Integration of sample storage and sample management for life science
US20060099567A1 (en) * 2004-04-08 2006-05-11 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20080176209A1 (en) * 2004-04-08 2008-07-24 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20080307117A1 (en) * 2004-04-08 2008-12-11 Judy Muller-Cohn Integration of sample storage and sample management for life science
US8900856B2 (en) 2004-04-08 2014-12-02 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20050276728A1 (en) * 2004-04-08 2005-12-15 Biomatrica, Inc. Integration of sample storage and sample management for life science
US9078426B2 (en) 2004-04-08 2015-07-14 Biomatrica, Inc. Integration of sample storage and sample management for life science
US20080268514A1 (en) * 2007-04-24 2008-10-30 Rolf Muller Sample storage for life science
WO2009009210A3 (en) * 2007-04-24 2009-09-24 Biomatrica, Inc. Sample storage for life science
US20110117647A1 (en) * 2008-03-21 2011-05-19 Fenwal, Inc. Red Blood Cell Storage Medium For Extended Storage
US8871434B2 (en) 2008-03-21 2014-10-28 Fenwal, Inc. Red blood cell storage medium for extended storage
US8968992B2 (en) 2008-03-21 2015-03-03 Fenwal, Inc. Red blood cell storage medium for extended storage
US20090239208A1 (en) * 2008-03-21 2009-09-24 Veronique Mayaudon Red Blood Cell Storage Medium For Extended Storage
WO2009120996A1 (en) * 2008-03-27 2009-10-01 Biolife Solutions, Inc. Mehtod, system, and apparatus for hypothermic collection, storage, transport and banking of birth tissue
WO2009121002A1 (en) * 2008-03-27 2009-10-01 Biolife Solutions, Inc. Materials and methods for hypothermic collection of whole blood
US20110152719A1 (en) * 2008-03-27 2011-06-23 Biolife Solutions, Inc. Materials and methods for hypothermic collection of whole blood
US8642255B2 (en) 2008-03-27 2014-02-04 Biolife Solutions, Inc. Materials and methods for hypothermic collection of whole blood
JP2015051986A (en) * 2008-12-05 2015-03-19 リニューロン・リミテッドReNeuron Limited Cellular compositions for use in therapy
JP2012510986A (en) * 2008-12-05 2012-05-17 リニューロン・リミテッド Therapeutic cell composition
WO2010064054A1 (en) * 2008-12-05 2010-06-10 Reneuron Limited Cellular compositions for use in therapy
US9265795B2 (en) 2008-12-05 2016-02-23 Reneuron Limited Cellular compositions for use in therapy
AU2009323841B2 (en) * 2008-12-05 2013-04-11 Reneuron Limited Cellular compositions for use in therapy
ES2343721A1 (en) * 2008-12-19 2010-08-06 Histocell, S.L. Cell transport system
US9410122B2 (en) 2009-02-06 2016-08-09 Reneuron Limited Treatment of limb ischemia
US9345857B2 (en) 2009-04-29 2016-05-24 Cardio3 Biosciences Injection catheter for delivering a therapeutic agent into a substrate
US10625051B2 (en) 2009-04-29 2020-04-21 Celyad S.A. Injection catheter for delivering a therapeutic agent into a substrate
US9943077B2 (en) 2009-10-23 2018-04-17 Fenwal, Inc. Methods for storing red blood cell products
US11864553B2 (en) 2009-10-23 2024-01-09 Fenwal, Inc. Methods and systems for providing red blood cell products with reduced plasma
US9409128B2 (en) 2009-10-23 2016-08-09 Fenwal, Inc. Methods for storing red blood cell products
US9845489B2 (en) 2010-07-26 2017-12-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA and proteins in saliva and other biological samples during shipping and storage at ambient temperatures
US9999217B2 (en) 2010-07-26 2018-06-19 Biomatrica, Inc. Compositions for stabilizing DNA, RNA, and proteins in blood and other biological samples during shipping and storage at ambient temperatures
US9376709B2 (en) 2010-07-26 2016-06-28 Biomatrica, Inc. Compositions for stabilizing DNA and RNA in blood and other biological samples during shipping and storage at ambient temperatures
US9725703B2 (en) 2012-12-20 2017-08-08 Biomatrica, Inc. Formulations and methods for stabilizing PCR reagents
US10064404B2 (en) 2014-06-10 2018-09-04 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US10772319B2 (en) 2014-06-10 2020-09-15 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US11672247B2 (en) 2014-06-10 2023-06-13 Biomatrica, Inc. Stabilization of thrombocytes at ambient temperatures
US10568317B2 (en) 2015-12-08 2020-02-25 Biomatrica, Inc. Reduction of erythrocyte sedimentation rate
US11116205B2 (en) 2015-12-08 2021-09-14 Biomatrica, Inc. Reduction of erythrocyte sedimentation rate
CN108477143A (en) * 2018-03-27 2018-09-04 福建三造血技术有限公司 A kind of cell transport holding liquid and transportation resources
CN114586768A (en) * 2020-12-03 2022-06-07 江苏齐氏生物科技有限公司 Liver tissue preservation solution and preservation method and application thereof

Similar Documents

Publication Publication Date Title
US20070048726A1 (en) Methods and Compositions for the Control of Molecular-Based Cell Death During Preservation of Cells, Tissues or Organs in a Gel-Like State
US6045990A (en) Inclusion of apoptotic regulators in solutions for cell storage at low temperature
US6632666B2 (en) Normothermic, hypothermic and cryopreservation maintenance and storage of cells, tissues and organs in gel-based media
US6921633B2 (en) Methods and compositions for the preservation of cells, tissues or organs in the vitreous state
YOUNIS et al. The effects of antifreeze peptide III (AFP) and insulin transferrin selenium (ITS) on cryopreservation of chimpanzee (Pan troglodytes) spermatozoa
Isachenko et al. Acrosomal status and mitochondrial activity of human spermatozoa vitrified with sucrose
US5635344A (en) Shipping medium for organ-derived cells
US20050106554A1 (en) Cryopreservation of pluripotent stem cells
US20090029340A1 (en) Cryoprotective Compositions and Methods of Using Same
Thuwanut et al. Cryopreservation of epididymal cat spermatozoa: effects of in vitro antioxidative enzymes supplementation and lipid peroxidation induction
Silva et al. Effect of coconut water and Braun-Collins solutions at different temperatures and incubation times on the morphology of goat preantral follicles preserved in vitro
Pichugin et al. Cryopreservation of rat hippocampal slices by vitrification
JP2008501320A (en) Cell preservation method
Tariq et al. Effect of carboxylated poly l-Lysine as a cryoprotectant on post-thaw quality and in vivo fertility of Nili Ravi buffalo (Bubalus bubalis) bull semen
Öztürk et al. The synergistic effect of trehalose and low concentrations of cryoprotectants can improve post-thaw ram sperm parameters
Bottrel et al. Cryoprotective effect of glutamine, taurine, and proline on post-thaw semen quality and DNA integrity of donkey spermatozoa
EL-RAEY et al. Effect of melatonin on buffalo bull sperm freezability, ultrastructure changes and fertilizing potentials
Neto et al. Effects of different freezing parameters on the morphology and viability of preantral follicles after cryopreservation of doe rabbit ovarian tissue
Tomalty et al. Chilling injury in human kidney tubule cells after subzero storage is not mitigated by antifreeze protein addition
Rocha et al. Evaluation of two different boar semen freezing protocols and their effects on semen quality after thawing
Cheema et al. Supplementation of Enzymatic and Non-enzymatic Antioxidants to the Extender improves Sperm Functionality during Storage at 4° C in Labrador Dog
Armitage Developments in corneal preservation
Datta et al. Development of a new method to preserve caprine cauda epididymal spermatozoa in-situ at-10 C with electrolyte free medium
Fisher et al. Cold and cryopreservation of monkey liver slices
Aktar et al. Effects of alpha lipoic acid supplementation on post-thaw quality of drone semen

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOLIFE SOLUTIONS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BAUST, JOHN M;VAN BUSKIRK, ROBERT;BAUST, JOHN G;AND OTHERS;REEL/FRAME:018441/0135

Effective date: 20061026

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION